New Stem Cell Research Offers First Glimpse of Early Human Development – SciTechDaily

Using a novel stem cell model, scientists have advanced our understanding of gastrulationa critical early stage of human developmentoffering new insights that could improve outcomes in pregnancy and the understanding of developmental disorders. The image above shows a blastoid, a stem cell model system that allows scientists to study the nuances of human gastrulation. Credit: Laboratory of Stem Cell Biology and Molecular Embryology at The Rockefeller University

Its one of lifes most defining momentsthat crucial step in embryonic development, when an indistinct ball of cells rearranges itself into the orderly three-layered structure that sets the stage for all to come. Known as gastrulation, this crucial process unfolds in the third week of human development. Gastrulation is the origin of our own individualization, the emergence of our axis, says Rockefellers Ali Brivanlou. It is the first moment that separates our heads from our behinds.

Observing the molecular underpinnings of this pivotal event would go a long way toward helping scientists prevent miscarriages and developmental disorders. But studying human gastrulation has proven both technologically difficult and ethically complicated, and thus current approaches have had limited success in expanding our understanding of early human development. Now Brivanlou and colleagues have demonstrated how a stem cell model system known as a blastoid can allow the study of the nuances of human gastrulation in the presence of pre-implantation extra-embryonic cell types. Their study, published in Stem Cell Reports, describes the scientific and clinical potential of this new platform.

Gastrulation was a tremendous black box. We had never seen ourselves at that stage, Brivanlou says. This moves us closer to understanding how we begin.

Prior to implantation, an embryo is a ball of about 250 cells organized as a blastocyst. This elusive ball of cells was difficult to study directly, so scientists developed blastoidsstem-cell-based blastocyst models. Blastoids can be cloned, experimentally manipulated, and programmed, allowing scientists to study identical blastoids over and over again.

The question was whether blastoids could gastrulate in vitro. Unlike a blastocyst in vivo, which rolls around in the uterus until it attaches to maternal tissue, blastoids were good at modeling the ball of cells from which life emerges, but it remained unclear whether this in vitro model could model later stages of human development. That is, until Brivanlou developed a platform to allow blastoids to attach in vitro, and thereby progress toward gastrulation.

We were then able to see epiblast symmetry breaking, marked by BRA expression, for the first time with the high molecular resolution, says Riccardo De Santis, a research associate in the Brivanlou lab and lead author on the study. This allowed us to start asking more detailed questions about the earliest moments of life.

With this unprecedented clarity, the team directly observed two key moments in gastrulation: the first epiblast symmetry-breaking event and the emergence of the molecular markers of the primitive streak and mesoderm upon in vitro attachment.

The primitive streak is a structure that marks the beginning of gastrulation and lays the foundation for the three primary layers of the embryo. One of those layers, the mesoderm, forms during gastrulation and gives rise to muscles, bones, and the circulatory system. The team discovered that, as early as seven days after attachment, they were already able to use molecular markers to detect the earliest signature of a nascent primitive streak and mesodermal cells.

To confirm their findings, the team also compared the blastoid results with data from in vitro attached human embryos and demonstrated that blastoids express the same genes in vitro that a regular embryo would at that stage in vivo, a strong demonstration of the power of blastoids as models for human embryonic development. Further highlighting the power of the labs in vitro attached blastoid system, the team then used it to demonstrate that pathways that regulate the rise of the primitive streak and mesoderm in vivo also regulate blastoids symmetry breaking in vitroall with nothing but stem-cell-derived blastoid models.

Along the way, the team also demonstrated that gastrulation in vitro can begin at day 12, earlier than once thought. This will change textbooks, Brivanlou says. Weve contributed to redefining the molecular signature and timing of the onset of gastrulation upon in vitro attachment.

The results demonstrate that blastoids, when combined with the Brivanlou labs unique attachment platform, are now capable of conveying insights into early human development that have long been inaccessible. De Santis envisions a future in which blastoid-based research leads to advancements in diagnosing and treating developmental disorders, or offers insights into potential causes of early miscarriages during gastrulation.

Many couples cant have babies because the embryo doesnt attach properly, and many miscarriages occur in the first few weeks of pregnancy, De Santis explains. We now have a model system that can help us understand the molecular mechanism that defines whether a pregnancy will be successful or not. In the near future, De Santis hopes to combine this method with machine learning to help predict pregnancy outcomes and the trajectories of developmental disorders by observing how model blastoids built with particular genetic makeups fare in vitro.

A better understanding of gastrulationand the ability to study it with a reliable model systemimpacts everything from survival of the fetus to autism to neurodegeneration.

Reference: The emergence of human gastrulation upon in vitro attachment by Riccardo De Santis, Eleni Rice, Gist Croft, Min Yang, Edwin A. Rosado-Olivieri and Ali H. Brivanlou, 14 December 2023, Stem Cell Reports. DOI: 10.1016/j.stemcr.2023.11.005

Visit link:
New Stem Cell Research Offers First Glimpse of Early Human Development - SciTechDaily

Mesenchymal stem cell based therapies for uveitis: a systematic review of preclinical studies | Eye – Nature.com

Wakefield D, Chang JH. Epidemiology of uveitis. Int Ophthalmol Clin. 2005;45:113.

Article PubMed Google Scholar

Miserocchi E, Fogliato G, Modorati G, Bandello F. Review on the worldwide epidemiology of uveitis. Eur J Ophthalmol. 2013;23:70517.

Article PubMed Google Scholar

Babu K, Mahendradas P. Medical management of uveitis - current trends. Indian J Ophthalmol. 2013;61:27783.

Article PubMed PubMed Central Google Scholar

Zhao PT, Zhang LJ, Shao H, Bai LL, Yu B, Su C, et al. Therapeutic effects of mesenchymal stem cells administered at later phase of recurrent experimental autoimmune uveitis. Int J Ophthalmol. 2016;9:13819.

PubMed PubMed Central Google Scholar

Li J, Qiu C, Zhang Z, Yuan W, Ge Z, Tan B, et al. Subretinal transplantation of human amniotic epithelial cells in the treatment of autoimmune uveitis in rats. Cell Transpl. 2018;27:150414.

Article Google Scholar

Zhang L, Zheng H, Shao H, Nian H, Zhang Y, Bai L, et al. Long-term therapeutic effects of mesenchymal stem cells compared to dexamethasone on recurrent experimental autoimmune uveitis of rats. Investig Ophthalmol Vis Sci. 2014;55:556171.

Article CAS Google Scholar

Margiana R, Markov A, Zekiy AO, Hamza MU, Al-Dabbagh KA, Al-Zubaidi SH, et al. Clinical application of mesenchymal stem cell in regenerative medicine: a narrative review. Stem Cell Res Ther. 2022;13:366.

Article CAS PubMed PubMed Central Google Scholar

Kanda P, Davis DR. Cellular mechanisms underlying cardiac engraftment of stem cells. Expert Opin Biol Ther. 2017;17:112743.

Article CAS PubMed Google Scholar

Levy O, Kuai R, Siren EMJ, Bhere D, Milton Y, Nissar N, et al. Shattering barriers toward clinically meaningful MSC therapies. Sci Adv. 2020;6:eaba6884.

Article CAS PubMed PubMed Central Google Scholar

Li Y, Ren X, Zhang Z, Duan Y, Li H, Chen S, et al. Effect of small extracellular vesicles derived from IL-10-overexpressing mesenchymal stem cells on experimental autoimmune uveitis. Stem Cell Res Ther. 2022;13:100.

Article CAS PubMed PubMed Central Google Scholar

Wei W, Ao Q, Wang X, Cao Y, Liu Y, Zheng SG, et al. Mesenchymal stem cell-derived exosomes: a promising biological tool in nanomedicine. Front Pharm. 2020;11:590470.

Article CAS Google Scholar

Liberati A, Altman DG, Tetzlaff J, Mulrow C, Gotzsche PC, Ioannidis JP, et al. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate healthcare interventions: explanation and elaboration. BMJ. 2009;339:b2700.

Article PubMed PubMed Central Google Scholar

Kung J, Chiappelli F, Cajulis OO, Avezova R, Kossan G, Chew L, et al. From systematic reviews to clinical recommendations for evidence-based health care: validation of Revised Assessment of Multiple Systematic Reviews (R-AMSTAR) for grading of clinical relevance. Open Dent J. 2010;4:8491.

PubMed PubMed Central Google Scholar

Hooijmans CR, Rovers MM, de Vries RB, Leenaars M, Ritskes-Hoitinga M, Langendam MW. SYRCLEs risk of bias tool for animal studies. BMC Med Res Methodol. 2014;14:43.

Article PubMed PubMed Central Google Scholar

McHugh ML. Interrater reliability: the kappa statistic. Biochem Med. 2012;22:27682.

Article Google Scholar

Zhang X, Ren X, Li G, Jiao C, Zhang L, Zhao S, et al. Mesenchymal stem cells ameliorate experimental autoimmune uveoretinitis by comprehensive modulation of systemic autoimmunity. Investig Ophthalmol Vis Sci. 2011;52:314352.

Article CAS Google Scholar

Tasso R, Ilengo C, Quarto R, Cancedda R, Caspi RR, Pennesi G. Mesenchymal stem cells induce functionally active T-regulatory lymphocytes in a paracrine fashion and ameliorate experimental autoimmune uveitis. Investig Ophthalmol Vis Sci. 2012;53:78693.

Article CAS Google Scholar

Qin Y, Chan AM, Chang YL, Matynia A, Kouris NA, Kimbrel EA, et al. Human embryonic stem cell-derived mesenchymal stromal cells decrease the development of severe experimental autoimmune Uveitis in B10.RIII Mice. Ocul Immunol Inflamm. 2018;26:122836.

Article CAS PubMed Google Scholar

Oh JY, Kim TW, Jeong HJ, Lee HJ, Ryu JS, Wee WR, et al. Intraperitoneal infusion of mesenchymal stem/stromal cells prevents experimental autoimmune uveitis in mice. Mediat Inflamm. 2014;2014:624640.

Article Google Scholar

Li G, Yuan L, Ren X, Nian H, Zhang L, Han ZC, et al. The effect of mesenchymal stem cells on dynamic changes of T cell subsets in experimental autoimmune uveoretinitis. Clin Exp Immunol. 2013;173:2837.

Article CAS PubMed PubMed Central Google Scholar

Lee HJ, Ko JH, Jeong HJ, Ko AY, Kim MK, Wee WR, et al. Mesenchymal stem/stromal cells protect against autoimmunity via CCL2-dependent recruitment of myeloid-derived suppressor cells. J Immunol. 2015;194:363445.

Article CAS PubMed Google Scholar

Ko JH, Lee HJ, Jeong HJ, Kim MK, Wee WR, Yoon SO, et al. Mesenchymal stem/stromal cells precondition lung monocytes/macrophages to produce tolerance against allo- and autoimmunity in the eye. Proc Natl Acad Sci USA. 2016;113:15863.

Article CAS PubMed Google Scholar

Chen X, Shao H, Zhi Y, Xiao Q, Su C, Dong L, et al. CD73 pathway contributes to the immunosuppressive ability of mesenchymal stem cells in intraocular autoimmune responses. Stem Cells Dev. 2016;25:33746.

Article PubMed Google Scholar

Mu Y, Xu W, Liu J, Wang Y, Chen J, Zhou Q. Mesenchymal stem cells moderate experimental autoimmune uveitis by dynamic regulating Th17 and Breg cells response. J Tissue Eng Regen Med. 2022;16:2635.

Article CAS PubMed Google Scholar

Bai L, Shao H, Wang H, Zhang Z, Su C, Dong L, et al. Effects of mesenchymal stem cell-derived exosomes on experimental autoimmune uveitis. Sci Rep. 2017;7:4323.

Article PubMed PubMed Central Google Scholar

Kang M, Choi JK, Jittayasothorn Y, Egwuagu CE. Interleukin 35-Producing exosomes suppress neuroinflammation and autoimmune uveitis. Front Immunol. 2020;11:1051.

Article CAS PubMed PubMed Central Google Scholar

Shigemoto-Kuroda T, Oh JY, Kim DK, Jeong HJ, Park SY, Lee HJ, et al. MSC-derived extracellular vesicles attenuate immune responses in two autoimmune murine models: Type 1 diabetes and uveoretinitis. Stem Cell Rep. 2017;8:121425.

Article CAS Google Scholar

Caspi RR. Experimental autoimmune uveoretinitis in the rat and mouse. Curr Protoc Immunol. 2003;15:15.16.120.

Google Scholar

Horai R, Caspi RR. Cytokines in autoimmune uveitis. J Interferon Cytokine Res. 2011;31:73344.

Article CAS PubMed PubMed Central Google Scholar

Foussat A, Gregoire S, Clerget-Chossat N, Terrada C, Asnagli H, Lemoine FM, et al. Regulatory T Cell therapy for uveitis: a new promising challenge. J Ocul Pharm Ther. 2017;33:27884.

Article CAS Google Scholar

Muthu S, Bapat A, Jain R, Jeyaraman N, Jeyaraman M. Exosomal therapy-a new frontier in regenerative medicine. Stem Cell Investig. 2021;8:7.

Article CAS PubMed PubMed Central Google Scholar

Nikfarjam S, Rezaie J, Zolbanin NM, Jafari R. Mesenchymal stem cell derived-exosomes: a modern approach in translational medicine. J Transl Med. 2020;18:449.

Article PubMed PubMed Central Google Scholar

Moller-Hansen M, Larsen AC, Toft PB, Lynggaard CD, Schwartz C, Bruunsgaard H, et al. Safety and feasibility of mesenchymal stem cell therapy in patients with aqueous deficient dry eye disease. Ocul Surf. 2021;19:4352.

Article PubMed Google Scholar

Calonge M, Perez I, Galindo S, Nieto-Miguel T, Lopez-Paniagua M, Fernandez I, et al. A proof-of-concept clinical trial using mesenchymal stem cells for the treatment of corneal epithelial stem cell deficiency. Transl Res. 2019;206:1840.

Article PubMed Google Scholar

Ozmert E, Arslan U. Management of retinitis pigmentosa by Whartons jelly derived mesenchymal stem cells: preliminary clinical results. Stem Cell Res Ther. 2020;11:25.

Article CAS PubMed PubMed Central Google Scholar

Galderisi U, Peluso G, Di Bernardo G. Clinical trials based on mesenchymal stromal cells are exponentially increasing: where are we in recent years? Stem Cell Rev Rep. 2022;18:2336.

Article PubMed Google Scholar

Siqueira RC, Messias A, Voltarelli JC, Scott IU, Jorge R. Intravitreal injection of autologous bone marrow-derived mononuclear cells for hereditary retinal dystrophy: a phase I trial. Retina. 2011;31:120714.

Article PubMed Google Scholar

Oner A, Gonen ZB, Sinim N, Cetin M, Ozkul Y. Subretinal adipose tissue-derived mesenchymal stem cell implantation in advanced stage retinitis pigmentosa: a phase I clinical safety study. Stem Cell Res Ther. 2016;7:178.

Article PubMed PubMed Central Google Scholar

Ezquer M, Urzua CA, Montecino S, Leal K, Conget P, Ezquer F. Intravitreal administration of multipotent mesenchymal stromal cells triggers a cytoprotective microenvironment in the retina of diabetic mice. Stem Cell Res Ther. 2016;7:42.

Article PubMed PubMed Central Google Scholar

Mastrolia I, Foppiani EM, Murgia A, Candini O, Samarelli AV, Grisendi G, et al. Challenges in clinical development of mesenchymal stromal/stem cells: concise review. Stem Cells Transl Med. 2019;8:113548.

Article PubMed PubMed Central Google Scholar

Kharbikar BN, Mohindra P, Desai TA. Biomaterials to enhance stem cell transplantation. Cell Stem Cell. 2022;29:692721.

Article CAS PubMed PubMed Central Google Scholar

Read the rest here:
Mesenchymal stem cell based therapies for uveitis: a systematic review of preclinical studies | Eye - Nature.com

Pluripotent positives in allogeneic stem cell therapies – BioProcess Insider

Ricardo Baptista, former chief technology officer at cell therapy developer Alder Therapeutics, told delegates The use of pluripotent stem cells is gaining traction when it comes to allogeneic stem cell therapies.

Baptista said there are several reasons for this and listed the benefits of using pluripotent stem cells. He discussed how pluripotent cells can be cultured in all systems, including 2D, 3D, static and dynamic. Additionally, Baptista said the lines can be edited [which equates] to the concept of a universal cell line and theoretically an unlimited choice of cells.

In turn, this means the therapies can be used off-the-shelf and target diseases with point-of-care therapy. Moreover, it is cell banks-based and there is the possibility to generate universal cells and the potential to leverage processing technologies from biopharma.

Baptista added there is an increased number of doses per lot and the costs of goods [is] spread across multiple doses, meaning the therapies are more easily accessible to a wider patient population.

Currently all approved chimeric antigen receptor (CAR) T-cell therapies are autologous. Autologous products are made by taking, reengineering, and reintroducing a patients own cells. Autologous methods of treatment usually have a low risk of rejection but are not always suitable for all patients because of the limitations in the quality and availability of the individuals cells.

Allogeneic therapies, however, can use cells or tissues from different individuals. As these are not personalized therapies, one advantage over autologous is the relative ease to mass-produce such products and thus, increase patient access. While allogeneic treatments could potentially treat more people, it has not yet fully reached commercialization due to the risk of rejection and immunosuppressive and matching measures required.

More here:
Pluripotent positives in allogeneic stem cell therapies - BioProcess Insider

Novel immune cell therapy may be a promising strategy for combating HBV infection – News-Medical.Net

Chronic infection with the hepatitis B virus (HBV) causes progressive liver problems, and eradication of the virus remains a formidable challenge. New research in FEBS Letters indicates that treatment that boosts the effects of immune cells called stem cell memory T cells (TSCMs) may be a promising strategy for combating HBV.

In the study, investigators identified TSCMs in patients with chronic HBV infection and analyzed their effects in a mouse model of HBV. After introducing TSCMs from patients into the mice, the immune cells differentiated into cytotoxic T cells and activated a strong response that resulted in the elimination of HBV-infected liver cells. Therefore, augmenting patients' TSCMs may help the immune system overcome the virus.

Our hepatitis model may reveal how TSCMs differentiate into cytotoxic T cells and uncover the fate of differentiated T cells. Such information could greatly advance future research on T cell-based therapies."

Hiromi Abe-Chayama, PhD, corresponding authorof Hiroshima University, Japan

Source:

Journal reference:

Abe-Chayama, H.,et al.(2024) Hepatitis B virus-specific human stem cell memory T cells differentiate into cytotoxic T cells and eradicate HBV-infected hepatocytes in mice. FEBS Letters. doi.org/10.1002/1873-3468.14842.

See the article here:
Novel immune cell therapy may be a promising strategy for combating HBV infection - News-Medical.Net

Tr1X Announces FDA Clearance of First Investigational New Drug Application for TRX103, an Allogeneic Regulatory T … – BioSpace

SAN DIEGO, April 10, 2024 /PRNewswire/ --Tr1X, Inc., anautoimmune and inflammatory disease cell therapy company focused on the development of novel allogeneic regulatory T cell therapies (Allo-Tregs) and allogeneic regulatory T cells expressing Chimeric Antigen Receptors (Allo-CAR Tregs), today announced the U.S. Food and Drug Administration (FDA) accepted the company's Investigational New Drug (IND) application for TRX103 for the prevention of Graft versus Host Disease (GvHD) in patients undergoing HLA-mismatched hematopoietic stem cell transplantation (HSCT). The company plans to initiate a Phase 1 study of TRX103, an investigational allogeneic off-the-shelf Tr1 Treg therapy, for this indication in the second quarter of 2024. Additionally, the company is on track to submit an IND for TRX103 for patients with refractory Crohn's disease in the third quarter of 2024.

"The FDA's clearance of our IND for TRX103, the first ever allogeneic engineered Tr1 regulatory T cell product, is an important milestone that could quickly provide us with proof-of-concept data while we continue to develop TRX103 for multiple autoimmune and inflammatory diseases, including Crohn's disease," said Maria Grazia Roncarolo M.D., Co-Founder, President and Head of R&D at Tr1X. "Donor-derived autologous Tr1 cells have shown clinical promise in improving immune reconstitution and reducing GvHD but have limited potential due to lack of feasibility and high cost. TRX103, an off-the-shelf product with unique biological properties compared to other Treg and CAR-T cell therapies, has the potential to reduce inflammation, suppress pathogenic cells, and reset the immune system. TRX103 is currently produced cost effectively at scale in a fully closed end-to-end system using a process that yields billions of cells in a single campaign. This should enable Tr1X to develop further pipeline candidates that address even larger patient populations with equally unmet medical needs."

"Allogeneic stem cell transplantation is the only curative treatment formany advanced blood cancers and genetic and acquired diseases.However, there remains a burden of morbidity and mortalityrelated to GvHD and its complications, including severe infections," said Monzr M. Al Malki, M.D., lead investigator of the Phase 1 study, Associate Professor in the Department of Hematology & Hematopoietic Cell Transplantation, and Director of the Unrelated Donor, Haploidentical and Cord Blood Transplant Programs at City of Hope National Medical Center. "As a result, innovative treatments are urgently needed. We look forward to starting this first-in-human trial to evaluate the safety, tolerability and clinical activity of these allogeneic Tr1 Treg cells and their potential to benefit patients in this setting."

About TRX103 TRX103 is an investigational allogeneic off-the-shelf engineered T cell product generated from CD4+ cells sourced from healthy donors. These donor-derived CD4+ cells are engineered to produce cells that mimic the function of Tr1 regulatory T cells. Tr1X is developing TRX103 for the treatment of several immune and inflammatory disorders. Multiple preclinical models of disease have shown TRX103 to be tolerable and effective and to have the potential to reset immune systems to a healthy state. TRX103 has the potential to overcome major limitations of current cell therapies for autoimmune diseases, which include limited persistence and side effects including cytokine release syndrome (CRS) and neurotoxicity.

About Tr1X Tr1X is a privately held biotechnology company focused on engineering cures for immune and inflammatory diseases. Founded by industry veterans, including the scientists behind the discovery of Tr1 cells, the company's pipeline of off-the-shelf allogeneic cell therapies is being developed for the treatment of and potential cure of autoimmune diseases with high unmet medical need. The company is backed by leading investors, including The Column Group, NEVA SGR and Alexandria Venture Investments, and has received additional grant support from the California Institute for Regenerative Medicine (CIRM). For more information visit http://www.tr1x.bio.

Investor Contact: Tr1X Investor Relations investors@tr1x.bio

Media Contact: Julie Normart jnormart@realchemistry.com

View original content to download multimedia:https://www.prnewswire.com/news-releases/tr1x-announces-fda-clearance-of-first-investigational-new-drug-application-for-trx103-an-allogeneic-regulatory-t-cell-therapy-to-treat-autoimmune-diseases-302112587.html

SOURCE Tr1X, Inc.

Follow this link:
Tr1X Announces FDA Clearance of First Investigational New Drug Application for TRX103, an Allogeneic Regulatory T ... - BioSpace

Stem cell treatment for spinal injury, and BRCA breakthrough | Podcasts – The Naked Scientists

Could an injection of stem cells grown from your own abdominal fat be the key to improving outcomes for people with spinal cord injuries? In an early stage trial in America, scientists have found that over two thirds of the small group of patients they treated showed improvements. They think that the stem cells are boosting the blood supply to the injured region of the spinal cord, and helping to control inflammation, which may lead to reduced scarring and better prospects for recovering some of the lost nerve connections. Mohamad Bydon is a neurosurgeon at the Mayo Clinic and led the new study

Mohamad - The historical teaching around spinal cord injury is you deliver surgery, you do physiotherapy, and that's basically it. And things haven't really advanced in a long, long time. So what we wanted to do was really impact the space and say, are there other treatments that we could add to augment the recovery, to aid the recovery, to improve the recovery?

Chris - And your intervention? What's the rationale behind what you're doing and how are you doing it?

Mohamad - So at a very high level, at a 30,000 foot view, the question becomes, what are the other things that we can add? And that's where we believe regenerative medicine will be a part of this paradigm. It's not going to be the only answer: you still need your surgery, you still need your physical therapy, there's other things like stimulation that are being discussed, but we believe regenerative therapy, specifically with stem cells, will be beneficial in helping to improve outcomes for patients.

Chris - So what stem cells? Where from and what do you do with them?

Mohamad - So stem cells are cells that can become a number of different things once they enter the body and they come from a number of different areas. Specifically in this study we used what are known as mesenchymal stem cells, adipose derived. Those words mean stem cells from your own fat, belly fat. I had a colleague who said to me recently, 'Who knew that belly fat could be so useful?' So, from your own belly fat, we remove that and expand the cells until we get to the right number of cells and then we proceed to reinject those cells once they're expanded and cultured into the spinal cord.

Chris - How many cells were you putting in once you'd grown them or expanded them and where exactly were they going? Were they going into the substance of the spinal cord or around it?

Mohamad - There were 100 million cells. Frankly, we need to work on dosing still, but a hundred million is the dose that we expand the cells to. Once we do that expansion, we proceed to inject it into the faecal sac. There's the substance of the spinal cord itself, and then there's a sac that surrounds the spinal cord - it's called the faecal sac or the dura mater - and that is a lining that surrounds the spinal cord. It also surrounds the brain. Inside that layer there's something called cerebrospinal fluid. So what we do is we put these cells inside the dura, into the fluid, and then the cells go to the area of highest injury and area of highest inflammation, which is the area of injury.

Chris - What do the cells look like? Are they still very much stem cells at this time when you're doing this? And then when they go to the areas of injury, is this only in people who've just had an injury or will they go to areas of injury that happened years ago?

Mohamad - Good question. The cells definitively are stem cells and there are certain markers and hallmark features that stem cells have. To your question on longevity, our current trial is in patients who've had their injury within a year. Many trials deliver therapy to patients who've had the injury right away; you had your injury yesterday, we're going to give you therapy today. This trial was not designed like that because some patients have natural improvement and so the earliest we injected any patient was at seven months. The latest that we injected any patient was at 22 months. Some of the patients that we injected out to 22 months had a very significant response. Now, we haven't done studies looking at longer, although now we're starting studies to look longer out. So what would it look like if we did patients after five years, ten years? What would that look like? Those are also things that we're evaluating and looking to treat.

Chris - Do you know for sure that the stem cells when you put them in actually go to the injury side or do they just go everywhere and some randomly crop up at the site of the injury? Have you actually followed them to see what happens to them and how long they persist for after you put them in?

Mohamad - We've done testing on this and we know that the cells go to the site of injury at the same time. The cells have an impact across the spinal cord and the brain and that's okay. The impact that we've seen has been positive or had no impact. So, we haven't seen it be negative. The cells themselves then work through a couple of different potential mechanisms once they get there. There's potentially a regenerative mechanism through the stem cells themselves regenerating that area, but the other potential mechanism is a vascular mechanism where the stem cells induce a more vascular area where scar tissue would normally be a very nonvascular area without blood vessels. Blood vessels are important because they deliver good nutrients, they take out bad nutrients, and so areas of injury tend to wall themselves off and lose their vasculature. These cells can be very helpful because they can reset the vasculature in those areas, allowing the areas to heal more properly.

Chris - For the patients, what were the outcomes like and in what ways did people improve in ways that you wouldn't have anticipated had they just been managed the way we normally, historically, have been managing spinal cord injuries?

Mohamad - What we looked at, in terms of safety, we found adverse events. Mostly, they were headaches or back pain that would improve over a few days. We never saw any significant or long-term side effects. On the effectiveness side, in terms of our secondary endpoint on effectiveness, what we found was that seven of the ten patients showed some improvement, three of them being very significant improvement, four of them being mild to moderate improvement, and the other three patients showed no improvement but did not get worse. Some patients who required a harness and multiple assistance to be able to bear weight and get up could now walk without that: they could walk on their own. Other patients had improvement in bowel and bladder function.

Chris - How do you know, though, that you didn't, just by chance, select people for this study who are that bit fitter? They're more likely to have a good outcome and, had they been just left to their own devices with the gold standard care they would otherwise have had, they would've ended up at the same endpoint?

Mohamad - This is a good question, and this is a question that we debated at length with the regulatory bodies. Most studies in this space treat patients right after the injury, in which case your question becomes very relevant. In our case, we waited. Most of the improvement after a spinal cord injury occurs within the first six months. Much less improvement occurs as you keep going over time, much, much less. The earliest we ever treated a patient was seven months and we had patients that we treated as late as 22 months and everybody had plateaued. Nobody was continuing their improvement. Remember, this is a phase 1 trial of ten patients. The definitive trial would be randomised controlled, which we're doing now, which is a phase 2 randomised controlled trial of best medical management versus our interventional therapy. But this is a signal and this is an important signal that will inform our future trials.

Read the original here:
Stem cell treatment for spinal injury, and BRCA breakthrough | Podcasts - The Naked Scientists

Human Neuron Model Paves the Way for New Alzheimer’s Therapies – Weill Cornell Medicine Newsroom

Weill Cornell Medicine scientists have developed an innovative human neuron model that robustly simulates the spread of tau protein aggregates in the braina process that drives cognitive decline in Alzheimer's disease and frontotemporal dementia. This new model has led to the identification of novel therapeutic targets that could potentially block tau spread.

The preclinical study, published April 5 in Cell, is a significant advancement in Alzheimer's disease research.

Dr. Li Gan. Credit: John Abbott

Currently no therapies can stop the spread of tau aggregates in the brains of patients with Alzheimers disease," said lead study author, Dr. Li Gan, director of the Helen and Robert Appel Alzheimers Disease Research Institute and the Burton P. and Judith B. Resnick Distinguished Professor in Neurodegenerative Diseases in theFeil Family Brain and Mind Research Instituteat Weill Cornell Medicine. Our human neuron model of tau spread overcomes the limitations of previous models and has unveiled potential targets for drug development that were previously unknown.

Human pluripotent stem cells can develop into any cell of the body and can be coaxed to become neurons to model brain diseases in a lab dish. However, it had been nearly impossible to model tau propagation in these young neurons, as tau propagation requires decades in aging brains.

Celeste Parra Bravo

Dr. Gans team used CRISPR technology to modify the genomes of human stem cells, prompting them to express forms of tau associated with diseased aging brains. "This model has been a game-changer, simulating tau spread in neurons within weeksa process that would typically take decades in the human brain," Dr. Gan said.

In their quest to halt tau propagation, Dr. Gan's team employed CRISPRi screening to disable 1,000 genes to ascertain their roles in tau spread. They discovered 500 genes that have a significant impact on tau abundance.

Dr. Shiaoching Gong

CRISPRi technology allowed us to use unbiased approaches to look for drug targets, not confined to what was previously reported by other scientists, said one of the lead study authors Celeste Parra Bravo, a neuroscience doctoral candidate in the Weill Cornell Graduate School of Medical Sciences working in the Gan lab.

One discovery includes the UFMylation cascade, a cellular process involving the attachment of a small protein named UFM1 to other proteins. This process's connection to tau spread was previously unknown. Post-mortem studies of brains from patients with Alzheimers disease found that UFMylation is altered, and the team also found in preclinical models that inhibition of the enzyme required for UFMylation blocks tau propagation in neurons.

We are particularly encouraged by the confirmation that inhibiting UFMylation blocked tau spread in both human neurons and mouse models, said paper co-author Dr. Shiaoching Gong, associate professor of research in neuroscience in the Appel Instituteat Weill Cornell Medicine.

Many Alzheimers disease treatments initially show promise in mouse models but do not succeed in clinical trials, Dr. Gan said. With the new human cell model, she is optimistic about the path ahead. "Our discoveries in human neurons open the door to developing new treatments that could truly make a difference for those suffering from this devastating disease."

Many Weill Cornell Medicine physicians and scientists maintain relationships and collaborate with external organizations to foster scientific innovation and provide expert guidance. The institution makes these disclosurespublic to ensure transparency. For this information, see profile forDr. Li Gan.

This research was supported in part by the National Institute of Neurological Disorders and Stroke, the National Institute on Aging and the National Institute of General Medical Sciences, all part of the National Institutes of Health, through grant numbers U54NS100717, R01AG072758, R01AG054214, R01AG074541, R25GM130494, and R01AG064239. Additional support was provided by the Rainwater Charitable Foundation and the JPB Foundation.

Excerpt from:
Human Neuron Model Paves the Way for New Alzheimer's Therapies - Weill Cornell Medicine Newsroom

Newark teenager suffering from a rare form of cancer who is having treatment funded by the NHS matched with stem … – LincsOnline

A teenager given a terminal diagnosis just before Christmas has now been told 95% of the cancer gone after experimental treatment.

Dan Evans, 18, is now looking to Israel for his next stage of treatment.

A major community fundraiser was launched to pay for the treatment after the teenager was told he had exhausted all avenues available on the NHS and to go home and make memories.

But money was not needed when the NHS agreed to fund the combination of drugs which had proved successful in trials for Dan.

And he is showing a positive response to treatment, having been told 95% of his cancer is gone.

Defying doctors and his diagnosis, a small part of the cancer is still visible on Dans neck but it has low activity.

Its still early days, but Dan certainly is on the right path now and its certainly a different path to the one he was on just a few months ago, said his dad, Mark.

As one of the final stages and vital to his recovery and to stay in remission once he gets to that stage, Dan is in need of a stem cell transplant. That process is due to start on May 27.

The NHS went on a world wide search for a matching donor. In such a short time, four stem cell donors have been found in Israel, however, the family needs to await confirmation on availability.

Dan had previously done the Ancestry test to discover more about his family history and roots, which showed him to be 1% connected to Israel.

Mark added: I am still concerned but I just feel that it is going to be OK, he is going to be fine.

After everything he has gone through, it just has to be fine, there is no other option really, it has to be fine,

Because there is still a little bit of cancer there, we are always going to be a little nervous, it is only natural.

The day he is finally in remission will be wonderful, absolutely wonderful and I cant wait.

Dan, a former pupil at Sir William Robertson Academy in Welbourn, was diagnosed with stage four Primary Mediastinal B Cell Lymphoma (PMBCL) in December 2022.

After several failed treatments, the family was told there were no more viable treatments available on the NHS and was told to take him home and make memories.

Instead, through research, Dan, who lives in Newark with his family, found a combination of drugs that had proved successful in trial, but not approved on the NHS.

His family set up a GoFundMe page with a 100,000 target and raised nearly half of their target amount in a short time before Dan was given the treatment for free on the NHS through compassionate use.

Dan had the first dose of treatment on January 26 and ever since, what once was a terminal diagnosis, has been improving and defying the rules of life and medicine.

Original post:
Newark teenager suffering from a rare form of cancer who is having treatment funded by the NHS matched with stem ... - LincsOnline

Dynamic molecular network analysis of iPSC-Purkinje cells differentiation delineates roles of ISG15 in SCA1 at the … – Nature.com

iPSC culture and differentiation to pan-neurons

SCA1-iPSCs and normal iPSCs were differentiated to pan-neurons87. SCA1-iPSCs and normal iPSCs were cultured in TeSR-E8 medium (STEMCELL Technologies, BC, Canada) with 10M Y27632 (253-00513, Wako, Osaka, Japan). After 24h, medium was changed to Stem Fit (AK02N, Ajinomoto, Tokyo, Japan) containing 5M SB431542 (13031, Cayman Chemical, Ann Arbor, MI, USA), 5M CHIR99021(13122, Cayman Chemical, Ann Arbor, MI, USA), and 5M dorsomorphin (044-33751, Wako, Osaka, Japan). After 5 days, iPSCs were dissociated with TrypLE Select (12563-011, Thermo Fisher Scientific, MA, USA). Neurospheres were then cultured in KBM medium (16050100, KHOJIN BIO, Saitama, Japan) with 20ng/mL Human-FGF-basic (100-18B, Peprotech, London, UK), 10ng/mL Recombinant Human LIF (NU0013-1, Nacalai, Kyoto, Japan), 10M Y27632 (253-00513, Wako, Osaka, Japan), 3M CHIR99021 (13122, Cayman Chemical, Ann Arbor, MI, USA), and 2M SB431542 (13031, Cayman Chemical, Ann Arbor, MI, USA) for 10 days. Finally, neurospheres were dissociated and seeded onto chambers coated with poly-L-ornithine (P3655, Sigma-Aldrich, St. Louis, MO, USA) and laminin (23016015, Thermo Fisher Scientific, Waltham, MA, USA), and cultured in DMEM/F12 (D6421, Sigma-Aldrich, St. Louis, MO, USA) supplemented with B27 (17504044, Thermo Fisher Scientific, Waltham, MA, USA), Glutamax (35050061, Thermo Fisher Scientific, Waltham, MA, USA), and penicillin/streptomycin (15140-122, Thermo Fisher Scientific, Waltham, MA, USA) for 14 days.

SCA1-iPSCs and normal iPSCs were differentiated to Purkinje cells88. To form EBs, iPSCs were dissociated with TrypLE Select (12563-011, Thermo Fisher Scientific, MA, USA), and 24,000 cells were aggregated by centrifugation at 200g for 2min in 96-well U-bottomed culture plates (650-180, Greiner, Kremsmnster, Austria) coated with Lipidure (CM5206, Nichiyu, Tokyo, Japan). Cells were cultured with gfCDM/insulin medium, 1:1 Iscoves modified Dulbeccos medium (12440053, Thermo Fisher Scientific, Waltham, MA, USA), and Hams F-12 (11765054, Thermo Fisher Scientific, Waltham, MA, USA) with 7g/mL insulin (I5500, Sigma-Aldrich, St. Louis, MO, USA); 1x chemically defined lipid concentrate (11905031, Thermo Fisher Scientific, Waltham, MA, USA); 15g/ml apo-transferrin (T4382, Sigma-Aldrich, St. Louis, MO, USA); 450M monothioglycerol (195-15791, Thermo Fisher Scientific, Waltham, MA, USA); 5mg/mL BSA (A7608, Sigma-Aldrich, St. Louis, MO, USA), Glutamax (35050061, Thermo Fisher Scientific, Waltham, MA, USA), and penicillin/streptomycin (15140-122, Thermo Fisher Scientific, Waltham, MA, USA); 20M Y-27632 (253-00513, Wako, Osaka, Japan); and 10M SB431542 (13031, Cayman Chemical, Ann Arbor, MI, USA). After 2 days, 50ng/mL recombinant human FGF2 (233-FB-025, R&D systems, MN, USA) was added to culture medium. After 21 days, EBs were transferred to 10cm Petri dishes (1020-100, Iwaki, Shizuoka, Japan) coated with Lipidure (CM5206, Nichiyu, Tokyo, Japan) and cultured for 14 days in Neurobasal/N2 medium, Neurobasal (21103-049, Thermo Fisher Scientific, Waltham, MA, USA) with N2 supplement (17502048, Thermo Fisher Scientific, Waltham, MA, USA), Glutamax (35050061, Thermo Fisher Scientific, Waltham, MA, USA), and penicillin/streptomycin (15140-122, Thermo Fisher Scientific, Waltham, MA, USA).

EBs were dissociated and cocultured with rhombic lip (RL) cells isolated from cerebellums of E14 Slc:ICR mice to induce differentiation into Purkinje cells. Briefly, RLs and EBs were dissociated with TrypLE Select (12563-011, Thermo Fisher Scientific, MA, USA) and cocultured in DMEM/F12 medium (11330032, Sigma-Aldrich, St. Louis, MO, USA) with 10% FBS, N2 supplement (17502048, Thermo Fisher Scientific, Waltham, MA, USA), and penicillin/streptomycin (15140-122, Thermo Fisher Scientific, Waltham, MA, USA). A total of 1.0106 cells at cell ratio = 1:10 (EB: RL) with 80L medium were seeded on chambers coated with poly-L-lysine (P1524-25MG, Sigma-Aldrich, St. Louis, MO, USA) and laminin (23016015, Thermo Fisher Scientific, Waltham, MA, USA). After incubation for 6h, DMEM/F-12 supplemented with N2 (17502048, Thermo Fisher Scientific, Waltham, MA, USA), 100g/mL BSA (A7608, Sigma-Aldrich, St. Louis, MO, USA), 50ng/mL human BDNF (248-BDB-010/CF, R&D systems, MN, USA), 50ng/mL human NT3 (267-N3-005/CF, R&D systems, MN, USA), and penicillin/streptomycin (15140-122, Thermo Fisher Scientific, Waltham, MA, USA) medium was added and cultured for 10 days.

EB-derived differentiating cells and RL-derived cells were cultured in cell culture insert dishes (140640, Thermo Fisher, Waltham, MA, USA) in which the two types of cells could be separated to avoid RL contamination of RNA-seq samples. The cell culture insert dish was coated with poly-L-lysine (P1524-25MG, Sigma-Aldrich, St. Louis, MO, USA) and laminin (23016015, Thermo Fisher Scientific, Waltham, MA, USA), embryonic body-derived cells were then seeded on the lower well, and RL-derived cells were seeded on a polycarbonate insert with a 4m pore. The culture medium was the same as described above.

To quantify the cell growth rate of SCA1-iPSCs and normal iPSCs, 30,000 iPSCs were seeded per well on Day 0. After 2, 4, 6, or 8 days, cells were collected, dissociated by 0.5 x TrypLE Select (12563-011, Thermo Fisher Scientific, MA, USA) and counted by using Burker-Turk hemocytometer. To quantify the size of iPSC-derived EBs, images of EBs were taken by microscope (IX70, Olympus) on day 1, 7, 14 and 21, and the 2D areas of EBs reflecting their 3D sizes were measured by ImageJ software (version 1.50, NIH, MD, USA).

AAV-HMGB1-EGFP or AAV-EGFP were infected into differentiated pan-neurons (MOI 2000). Twelve days after AAV infection, cells were fixed with 1% paraformaldehyde in PBS for 30min. After blocking in PBS containing 10% FBS for 30min, cells were stained with the following primary and secondary antibodies: mouse anti-III-tubulin 1 (1:2000 for 16h at 4C, #T8660 Sigma-Aldrich, St. Louis, MO, USA), rabbit anti-PSD95 (1:1000 for 16h at 4C, 3409, Cell Signaling Technology, Danvers, MA, USA), donkey anti-mouse IgG Alexa 488-conjugated (1:600 for 1h at room temperature, #715-545-150, Jackson ImmunoResearch Laboratories, West Grove, PA, USA), and donkey anti-rabbit IgG Alexa488-conjugated (1:1000 for 1h at room temperature, A-21206, Thermo Fisher Scientific). The dendritic spine was assessed after acquisition of image by confocal microscopy (FV1200 laser scanning microscope, Olympus, Tokyo, Japan). We used x40 objective lens (UPLSAPO40X2 (NA:0.95)). The dendritic spine density was measured by ImageJ software (version 1.50, NIH, MD, USA). After calibration of scale information, dendrite (Tuj-1-immunostained image) were manually traced and calculate the dendrite length in observing image window. Next, the spine (PSD95-immunostained dots) was counted, and the dendritic spine density was defined as the number of spines in 1m length of a dendrite, calculated by dividing a spine number in one dendrite by the length of it.

Differentiated Purkinje cells were fixed with 1% paraformaldehyde in PBS for 30min. Cells were incubated with 10% FBS followed by incubation with primary and secondary antibodies as follows: mouse anti-Calbindin (1:2000 for 16h at 4C, C9848, Sigma-Aldrich, St. Louis, MO, USA) and donkey anti-mouse IgG Alexa 488-conjugated (1:1000 for 1h at room temperature, A-21202, Thermo Fisher Scientific).

Mice brain tissues at different timepoints (P0, P28, P91, and P392) were fixed with 4% paraformaldehyde in 0.1M phosphate buffer for 12h and embedded in paraffin. Sagittal sections were deparaffinized in xylene and rehydrated in ethanol. For antigen retrieval, sections were incubated in Tris-EDTA solution pH 9.0 (100mM Tris-base and 10mM EDTA) at 121C for 15min. Human brain paraffin sections of SCA1 patients or control sections were also processed. Sections were then incubated with 0.5% triton-X 100 in PBS for 20min to perform permeabilization. Next, sections were incubated with 10% FBS in PBS for 30min and were incubated with primary and secondary antibodies as follows: rabbit anti-ISG15 (1:100 for 16h at 4C, HPA004627, Sigma-Aldrich, St. Louis, MO, USA), mouse anti-Calbindin (1:2000 for 16h at 4C, C9848, Sigma-Aldrich, St. Louis, MO, USA), mouse anti-Atxn1 (1:100 for 16h at 4C, MABN37, Millipore, Burlington, MA, USA), mouse anti-Ub (1:100 for 16h at 4C, #3936, Cell Signaling technology, Danvers, MA, USA), donkey anti-mouse IgG Alexa 488-conjugated (1:600 for 1h, #715-545-150, Jackson ImmunoResearch Laboratories, West Grove, PA, USA), and donkey anti-rabbit IgG Cy3-conjugated (1:600 for 1h, #711-165-152, Jackson ImmunoResearch Laboratories, West Grove, PA, USA). Nuclei were stained with DAPI (0.2g/mL in PBS, D523, DOJINDO Laboratories, Kumamoto, Japan). Z-stacked images (0.5m interval x 5 slices) were acquired from cerebellar cortex (Lobule IV/V) using a confocal microscope (FV1200IXGP44, Olympus, Tokyo, Japan) and a super-resolution microscope (LSM980 with Airyscan 2, Zeiss, Oberkochen, Germany). Signal intensities were measured using ImageJ software.

Male mice brain tissues at different time points (P0, P28, P91, and P392) were homogenized using the BioMasher II (#893062, Nippi, Tokyo, Japan) with RIPA buffer (10mM Tris-HCl pH 7.5, 150mM NaCl, 1mM EDTA, 1% Triton-X 100, 0.1% SDS, 0.1% DOC, and 1:250 volume Protease Inhibitor Cocktail (#539134, Calbiochem, San Diego, CA, USA)). Homogenates were centrifuged at 12,000g for 10min, and supernatants were added to equal volumes of sample buffer (0.1M Tris-HCl pH 6.8, 4% SDS, 20% glycerol, 0.05% BPB, and 12% -mercaptoethanol) and boiled at 100C for 10min. Samples were subjected to SDS-PAGE and transferred onto PVDF membrane. After blocking the membranes with 5% skim milk in TBST (20mM Tris-HCl pH 7.5, 150mM NaCl, 0.05% Tween-20) for 1h, membranes were incubated with primary and secondary antibodies as follows: rabbit anti-ISG15 (1:1000 for 3h at room temperature, HPA004627, Sigma-Aldrich, St. Louis, MO, USA), mouse anti-GAPDH (1:3000 for 16h at 4C, MAB374, Merck, Darmstadt, Germany), mouse anti-Atxn1 (1:1000 for 3h at room temperature, MABN37, Millipore, Burlington, MA, USA), mouse anti-Ub (1:1000 for 16h at 4C, #3936, Cell Signaling Technology, Danvers, MA, USA), mouse anti-Myc (1:3000 for 1h at room temperature, M047-3, MBL, Aichi, Japan), rabbit anti-FLAG (1:3000 for 1h at room temperature, F7425, Sigma, St. Louis, MO, USA), sheep anti-mouse IgG HRP conjugated (1:3000 for 1h, NA931, Cytiva, Tokyo, Japan), and rabbit anti-IgG HRP conjugated (1:3000 for 1h, NA934, Cytiva, Tokyo, Japan). Proteins were detected using Amersham ECL select (RPN2235, Cytiva, Tokyo, Japan) on an Image-Quant luminescence image analyzer LAS500 (Cytiva, Tokyo, Japan). Signal intensities were measured using ImageJ software.

SCA1-iPSCs and normal iPSCs were collected and homogenized in 350L RNA RLT buffer (Qiagen)/0.01% 2-mercaptoethanol (Wako, Tokyo, Japan). Total RNA was purified with RNeasy mini kit (Qiagen). To eliminate genomic DNA contamination, on-column DNA digestion was conducted for each sample with DNase I (Qiagen). Prepared RNA samples were subjected to a HiSeq-based RNA-seq by TAKARA (700 million bp reads).

Gene expression profiles of each sample were evaluated by the number of short reads that were mapped onto gene coding sequences in the reference human genome assembly hg38. Differential expression genes were analyzed with DESeq234. Log2FC (Fold Change) between SCA1-derived and normal cells was calculated by DESeq2, and the difference of gene expression was determined at | Log2FC|>0.5.

To generate the pathological network based on PPI, UniProt accession numbers were added to genes identified in RNA-seq-based gene expression analysis. The pathological PPI network was constructed by connecting genes using the integrated database of the Genome Network Project (GNP) (https://cell-innovation.nig.ac.jp/GNP/index_e.html), which includes BIND, BioGrid (http://www.thebiogrid.org/), HPRD, IntAct (http://www.ebi.ac.uk/intact/site/index.jsf), and MINT. A database of GNP-collected information was created on the Supercomputer System available at the Human Genome Center of the University of Tokyo.

To create a static molecular network, statistically significantly changed molecules were connected at two neighboring time points based on interactions in the PPI database (an integrated database collected by GNP, which includes BIND, BioGrid (http://www.thebiogrid.org/), HPRD, IntAct (http://www.ebi.ac.uk/intact/site/index.jsf), and MINT), without considering their cause-result relationships. Each network starting from a changed molecule was expanded step-by-step from one-hop (directly linked) to six extra connections, and the degree of significance at each expansion step was evaluated by calculating the z-score of their ratio of changed nodes.

To gain insights into the dynamics of the pathological molecular network, a PPI-based chronological molecular network was constructed by connecting two proteins between the two neighboring time points using the integrated PPI database.

To estimate the impact of a significantly differentially expressed gene to the future time point, a gene in a certain time point was connected to a set of genes in the next time point based on the PPI database (an integrated database collected by GNP, which includes BIND, BioGrid (http://www.thebiogrid.org/), HPRD, IntAct (http://www.ebi.ac.uk/intact/site/index.jsf), and MINT) and defined as downstream genes. The magnitude of impact of a gene at the first time point (defined as r) was calculated by the ratio of downstream genes that were significantly changed in mRNA expression levels to all genes at the second time point. The selection of genes was performed based on comparison of impact of a specific gene (specific impact) and total impact at the second time point. The statistical significance of the impact was examined using two-tailed Fishers exact test with post-hoc Benjamini-Hochberg procedure (adjusted p-value<0.05, red dots). The statistical significance of mRNA expression change was examined using log2FC between SCA1 and normal cells (|Log2FC|> 0.5, blue dots). A digraph was created from a significantly differentially expressed gene at the original time point (iPSCs) to significant genes at the end point (Purkinje cells) via significant genes at the intermediate time point based on the impact analysis. The digraph predicts the original gene whose change at the initial time point leads to molecular changes at the final time point.

To select cytokine-relevant genes, Gene Ontology (GO) enrichment analysis was performed using clusterProfiler89 package in R. A list of genes included in a selected pathway was used as input of enrichGO function of clusterProfiler. From all the enrichment results, GO terms related to cytokine were selected to extract a list of cytokine-relevant genes. The GO terms related to cytokine were searched by keyword cytokine, and thereafter terms that were not related to cytokine such as cytokinesis were excluded.

A search for transcriptomic studies of SCA1 in Homo sapiens and Mus musculus in the NCBI Sequence Read Archive (SRA; https://www.ncbi.nlm.nih.gov/sra) was performed on August 7, 2023 using the keywords spinocerebellar ataxia type 1 and SCA1. Eight studies (PRJNA305316, PRJNA422988, PRJNA472147, PRJNA472754, PRJNA503578, PRJNA688073, PRJNA871289, and PRJNA903078) that include RNA-seq data and raw sequence information generated from cerebellar tissues of SCA1 mouse models were found, while no studies with human RNA-seq data were found.

Raw sequence data were downloaded from NCBI SRA using the SRA Toolkit (https://github.com/ncbi/sra-tools) and mapped to the M. musculus genome assembly GRCm38/mm10 using HISAT2. Gene expression was calculated using the featureCounts function from Subread v1.5.2. Gene expression differences between the SCA1 and control groups were tested using Welchs t-test. Gene expression changes with a p-value0.05 were considered significant.

RNA was isolated from iPSCs, EBs and Purkinje cells with TRIzol RNA Isolation Reagents (15596026, Thermo Fisher Scientific, MA, USA). Reverse transcription was performed by using the SuperScript VILO cDNA Synthesis kit (11754-250, Invitrogen, Carlsbad, CA, USA). Quantitative PCR analyses were performed with the 7500 Real-Time PCR System (Applied Biosystems, Foster City, CA, USA) by using THUNDERBIRD SYBR qPCR Mix (QPS-201, TOYOBO, Osaka, Japan) and assessed by the standard curve method. The primer sequences were:

ISG15, forward primer: 5-CGCAGATCACCCAGAAGATCG-3 and reverse primer: 5- TTCGTCGCATTTGTCCACCA-3

UbE2L6, forward primer: 5-GTGGCGAAAGAGCTGGAGAG-3 and reverse primer: 5 -ACACTGTCTGCTGGTGGAGTTC- 3

ARIH1, forward primer: 5-CAGGAGGAGGATTACCGCTAC-3 and reverse primer: 5-CTCCCGGATACATTCCACCA-3

GAPDH, forward primer: 5-AGATCATCAGCAATGCCTCCTG-3 and reverse primer: 5-ATGGCATGGACTGTGGTCATG-3

PCR conditions for amplification were 40 cycles of 95C for 1min for enzyme activation, 95C for 15sec for denaturation, and 60C for 1min for extension. The expression levels of ISG15, UbE2L6 and ARIH1 were corrected by GAPDH.

Frozen mouse brains (male, P28) were lysed with TNE buffer (10mM Tris-HCL (pH 7.5), 150mM NaCl, 1mM EDTA, and 1% NP-40) and collected by centrifugation (15,000g10min). Aliquots (100g protein in cerebellar tissue lysate) were then incubated 1h with a 50% slurry of protein G-sepharose beads. After centrifugation (2000g3min), the supernatants were incubated with 1g rabbit anti-ISG15 antibody (aHPA004627, Sigma-Aldrich, St. Louis, MO, USA) overnight at 4C. Reactants were then incubated with Protein G-sepharose beads for 4h, washed with TNE buffer, and eluted by sample buffer. For double-precipitation, samples were incubated with 2g biotin-labeled mouse anti-Ub (#3936, Cell Signaling technology, Danvers, MA, USA) overnight at 4C, followed by incubation with streptavidin beads (TrueBlot(R) Streptavidin Magnetic Beads, S000-18-5, Rockland, Pottstown, PA, USA). The collected samples were further incubated with 2g mouse anti-Atxn1 (MABN37, Millipore, Burlington, MA, USA) overnight at 4C. Then, reactants were incubated with Protein G-sepharose beads for 4h, washed with TNE buffer, and eluted in sample buffer.

HeLa cells were seeded at a density of 4105 cells/well in a 6-well plate (3516, Corning, Glendale, AZ, USA) and transfected with 30 pmol human ISG15-siRNA (sc-43869, Santa Cruz Biotechnology, Dallas, TX, USA) or scrambled siRNA (SR30004, OriGene, Rockville, MD, USA) using 4L Lipofectamine RNAiMAX (13778-075, Thermo Fisher Scientific, Waltham, MA, USA). At 24h after siRNA transfection, 2.5g myc-Ataxin1-33Q, myc-Ataxin1-86Q, or FLAG-Ku70 plasmid was transfected using 5L Lipofectamine 2000 (11668-019, Thermo Fisher Scientific, Waltham, MA, USA). At 48h after siRNA transfection, 100nM Bafilomycin A1 (BVT-0252-C100, AdipoGen, Liestal, Basel-Landschaft, Switzerland) or 5M MG132 (139-18451 Wako, Osaka, Japan) was added to the culture medium in order to inhibit autophagy or proteasome-dependent protein degradation, respectively. At 49h, 100g/mL cycloheximide (033-20993, Wako, Osaka, Japan) was added to the culture medium in order to inhibit protein synthesis. The cells were collected at 0, 6, 12, and 24h after addition of cycloheximide, lysed with RIPA buffer (10mM Tris-HCl pH 7.5, 150mM NaCl, 1mM EDTA, 1% Triton-X 100, 0.1% SDS, 0.1% DOC, and 1:250 volume Protease Inhibitor Cocktail (#539134, Calbiochem, San Diego, CA, USA)), and centrifuged at 12,000g for 10min. The supernatants were mixed with equal volumes of sample buffer (0.1M Tris-HCl pH6.8, 4% SDS, 20% glycerol, 0.05% BPB, and 12% -mercaptoethanol), boiled at 100C for 10min, and subjected to SDS-PAGE.

Mutant Atxn1-KI mice (Sca1154Q/2Q mice) were generously gifted by Prof. Huda Y. Zoghbi (Baylor College of Medicine, TX, USA)39. The backcrossed strain with C57BL/6 mice were further crossed with C57BL/6 female mice more than 10 times in our laboratory. The number of CAG repeats was checked by fragment analysis using the following primers: forward (5CACCAGTGCAGTAGCCTCAG3, labeled with 6-carboxyfluorescein) and reverse (5AGCTCTGTGGAGAGCTGGAA3). Mice were maintained under suitable humidity (around 50%) at 22C with a 12h light-dark cycle. We have complied with all relevant ethical regulations for animal use.

Cerebellar specimens collected at autopsy from three SCA1 patients and three control patients without neurological disorders (lung cancer, leukemia, and cholangiocarcinoma) were used. The details of the SCA1 patients (51-year-old female, 54-year-old female, and 50-year-old male) were described previously90,91,92. Their CAG repeat expansion in the Atxn1 gene was confirmed by PCR and their numbers of CAG repeats were around 50, although the exact numbers were not determined by fragment analysis or Sanger sequencing. Human plasma samples were acquired from SCA1 patients with a PCR-based genetic diagnosis or control patients without neurological disorders. Essential information about the SCA1 patients and controls is shown in Fig.8D. Other clinical information is not linked with samples according to ethics regulations. All ethical regulations to human research participants were followed.

In total, 100 L of human plasma samples that had been diluted 2-fold with saline (OTSUKA normal saline 20mL, Otsuka Pharmaceutical Factory, Tokushima, Japan) was added to a 96-well plate precoated with an anti-ISG15 antibody (#CY-8085, CircuLex Human ISG15 ELISA Kit, MBL, Tokyo, Japan) and incubated for 16h at 4C. Plates were washed and subsequently incubated with a peroxidase-conjugated anti-ISG15 antibody (Atlas Antibodies, HPA004627-100UL, Bromma, Sweden) for 2h at room temperature. For detection, Substrate Reagent (#CY-8085, CircuLex Human ISG15 ELISA Kit, MBL, Tokyo, Japan) was added to each well. The reaction was terminated with Stop Solution (#CY-8085, CircuLex Human ISG15 ELISA Kit, MBL, Tokyo, Japan), and absorbance at 450nm was measured on a microplate reader (SPARK 10M, TECAN, Grodig, Austria). A standard curve was generated using 0, 1.5, 3, 6, and 12ng/mL Recombinant Human ISG15 (UL-601-500, R&D Systems, Minneapolis, MN, USA) diluted with Sample Diluent (326078738, HMGB1 ELISA Kit Exp, Shino-test, Tokyo, Japan).

We analyzed three iPSC lines derived from two SCA1 patients for RNA-seq-based gene expression analysis. For meta-analysis using SCA1 model mice, we collected RNA-seq data from 3 to 17 mice in each time point from NCBI SRA database. Statistical analyses for biological experiments were performed using Graphpad Prism 8. Biological data following a normal distribution are presented as the meanSEM, with Tukeys HSD test or Dunnetts test for multiple group comparisons or with Welchs t-test for two group comparisons. The distribution of observed data was depicted with box plots, with the data also plotted as dots. Box plots show the medians, quartiles, and whiskers, which represent data outside the 25th75th percentile range. To obtain each data, we performed biologically independent experiments. The number of samples was indicated in each figure and figure legends.

This study was performed in strict accordance with the recommendations of the Guide for the Care and Use of Laboratory Animals of the Japanese Government and National Institutes of Health. All experiments were approved by the Committees on Gene Recombination Experiments, Human Ethics, and Animal Experiments of the Tokyo Medical and Dental University (G2018-082C3, 2014-5-3, and A2021-211A). Human samples including post-mortem brains were provided with informed consent and their use was approved by the Committees on Human Ethics (O2020-002-03).

Further information on research design is available in theNature Portfolio Reporting Summary linked to this article.

Read more here:
Dynamic molecular network analysis of iPSC-Purkinje cells differentiation delineates roles of ISG15 in SCA1 at the ... - Nature.com

ABVC BioPharma Executes a Global Licensing Term Sheet for Oncology/Hematology Products, Expecting Licensing Income of $55M and Royalties of up to $50M

FREMONT, CA, April 10, 2024 (GLOBE NEWSWIRE) -- via NewMediaWire -- ABVC BioPharma, Inc. (NASDAQ: ABVC) ("Company"), a clinical-stage biopharmaceutical company developing therapeutic solutions in ophthalmology, CNS (central nervous systems), and Oncology/Hematology, announced today that the Company together with its affiliates BioLite, Inc., and Rgene Corporation entered into a term sheet with OncoX BioPharma, Inc. (OncoX) for the Company's Oncology/Hematology pipeline(the "Licensed Products"). Subject to negotiation and execution of the definitive agreement, this license would cover the Licensed Products' clinical trial, registration, manufacturing, supply, and distribution rights. ABVC is set to receive $50,000,000 as licensing fees in the form of Cash/Shares within 30 days of execution of the Definitive Agreement, with an additional milestone payment of $5,000,000 in cash after the first fundraising round and Royalties of 5% of net sales, up to $50,000,000, after the launch of the Licensed Products.

See the rest here:
ABVC BioPharma Executes a Global Licensing Term Sheet for Oncology/Hematology Products, Expecting Licensing Income of $55M and Royalties of up to $50M