Treating a type 2 diabetic patient with impaired pancreatic islet function by personalized endoderm stem cell-derived … – Nature.com

Type 2 diabetes (T2D) typically starts with insulin resistance in peripheral tissues and proceeds with gradual loss of islet function due to the reduction in -cell mass or dedifferentiation of cells1,2. More than 30% of T2D patients eventually rely on exogenous insulin treatment. Cadaveric islet transplantation is an effective treatment for insulin-dependent diabetes3,4. Notably, improved metabolic control after islet transplantation is associated with better kidney allograft function and long-term survival5,6. However, the application of islet transplantation is severely hampered due to the critical shortage of donor organs.

The pancreatic progenitor (PP) cells or islet tissues, generated from human pluripotent stem cells (hPSCs), have been shown to survive, function and reverse hyperglycemia in diabetic animal models7,8,9. In addition, a recent clinical trial has shown that, when subcutaneously implanted into T1D patients, the hPSC-derived pancreatic endodermal cells encapsulated with non-immunoprotective devices were able to further mature into meal-responsive -like cells and secrete insulin, albeit at the levels insufficient to achieve the independence of exogenous insulin10,11. Nevertheless, clinical applications of hPSC-derived cells are undermined by the complicated differentiation processes and the risk of having residual undifferentiated cells that may form teratomas in vivo. Recent studies have focused on identifying intermediate stem cell types, including the non-tumorigenic human endoderm stem cells (EnSCs)12, which appear to be more suitable as precursors for large-scale generation of islet cells.

Here, we report the intrahepatic implantation of islet tissue (E-islets) differentiated in vitro from autologous EnSCs in a T2D patient who had impaired insulin secretion. This is a pilot study of an investigator-initiated trial designed to investigate the safety and efficacy of E-islets for the treatment of insulin-dependent diabetic patients (Fig. 1a). The patient was a 59-year-old man with a 25-year history of T2D who developed end-stage diabetic nephropathy and underwent kidney transplantation in June of 2017 and displayed poor glycemic control since November of 2019, characterized by blood glucose level ranging from 3.6614.60mmol/L, mean amplitude of glycemic excursion (MAGE) of 5.54mmol/L, the time-in-the-tight-target-range (TITR, 3.97.8mM) of 56.7%, with daily hyperglycemic events (> 10.0mmol/L) of 0.7/d and hypoglycemic events (< 3.9mmol/L) of 0.3/d (Supplementary Table S1). Due to the major concerns of hypoglycemia and the detrimental effect of poor glycemic control on the long-term survival of the donor kidney, the patient agreed to pursue transplantation with autologous E-islets.

a Brief scheme of major procedures involved in the generation and quality control of E-islets and the safety/effectiveness evaluations of E-islet transplantation. bd E-islets reverse hyperglycemia in STZ-induced diabetic immunocompromised mice. Schematic illustration of kidney capsule transplantation of E-islets (b). Fasting blood glucose dynamics (blue line: sham group; red line: E-islet-transplanted group, c). Secretion of human C-peptide after fasting and 30min following an i.p. glucose bolus on days 90 and 180 post transplantation (d). eg Immunogenicity of E-islets in humanized mice. Schematic illustration of the syngeneic and allogeneic kidney capsule transplantation of patient-specific E-islets into the NCG-hIL15 diabetic mice humanized with the patients and a volunteers PBMCs (e). Fasting blood glucose dynamics (blue line represents the control group with the patient E-islets transplanted into three diabetic mice humanized with the volunteers PBMCs; red line represents the group with the patient E-islets transplanted into three diabetic mice humanized with the patients PBMCs, f). Secretion of human C-peptide after fasting and 30min following an i.p. glucose bolus on days 7 and 14 post E-islet transplantation (U.D. undetectable, g). h Clinical measurements of TITR, TIR and HbA1c, and the insulin dosage during 116 weeks. i Continuous interstitial glucose fluctuations derived from the CGM measurements at weeks 52 and 105 compared with pre-surgery levels. jl Serum levels of fasting and meal-stimulated circulating glucose (j), C-peptide (k) and insulin (l) from MMTT assays.

E-islets were generated from the autologous EnSCs that are established under the culture condition modified from our previous reports12 (see details in Supplementary methods), through two intermediate stages under GMP conditions. The morphology, purity, viability and microorganism contamination of EnSC-derived PPs, endocrine progenitor cells and E-islets were proven to meet the release criteria (Supplementary Figs. S1S3 and Table S6). E-islets displayed similar morphology (Supplementary Fig. S3a), endocrine cell composition (Supplementary Fig. S3b, c, e, f), gene expression patterns (Supplementary Fig. S3df) and in vitro functionality (Supplementary Fig. S3g) to human cadaveric islets, and showed functional efficacy in Streptozotocin (STZ)-induced diabetic mouse (Fig. 1bd) and monkey (Supplementary Fig. S4) models. The nontarget hepatic or intestinal lineages, when examined by either scRNA-seq (Supplementary Fig. S3f) or FACS (Supplementary Fig. S3h), were not detected. Neither tumor formation nor cystic/ductal structures that indicate cell proliferation were detected in the immunocompromised animals transplanted with either EnSCs or E-islets during the experiments (Supplementary Table S3). The patient-specific E-islets survived and functioned under the kidney capsules of the diabetic immunocompromised mice humanized with patients own PBMCs, but rejected by the ones humanized with PBMCs from an unrelated volunteer (Fig. 1eg; Supplementary Fig. S5), which suggests that patients immune system likely tolerates the autologous E-islets.

The patient underwent a percutaneous transhepatic portal vein transplantation with 1.2 million IEQs of E-islets delivered, conforming to the regulatory guidance from the clinical islet transplantation registration. At designated visits, examinations of endocrine function and diabetes-specific parameters by mixed-meal tolerance test (MMTT) were performed at baseline, 4, 8, 12, 16, 20, 24, 36 and 48 weeks and thereafter at specified time points (Supplementary Fig. S6a). The glycemic control of the patient was measured with a 24-h real-time continuous glucose monitoring system (CGM).

During the 116-week follow-up period, no tumor formation was detected either by MRI on the upper abdomen or by the measurements of serum tumor-related antigen markers. The treatment-emergent adverse events included: (1) temporary abdominal distension and loss of appetite within 48 weeks, relieved with methionyltrichloride; (2) restorable weight loss < 5% (from 80kg to 76kg).

The three major clinical outcomes, the glycemic targets, the reduction of exogenous insulin and the levels of fasting and meal-stimulated circulating C-peptide/insulin were monitored throughout the first 116 weeks (Supplementary Tables S4, S5). Marked changes in the patients glycemic control were observed as early as week 2 post transplantation, as the MAGE declined from 5.50mmol/L to 3.60mmol/L, and the TITR increased rapidly from 56.7% to 77.8% (Fig. 1h; Supplementary Table S1). Over the same period, the time-above-range (TAR) decreased by 55% from baseline, while the events of severe hyperglycemia (> 13.9mM) and hypoglycemia (<3.9mM) completely disappeared (Supplementary Fig. S7a, b and Table S1). During the period between weeks 4 and 12, a significant reduction in ambulatory mean glucose fluctuations (from 5.50 to 2.6mmol/L) (Supplementary Table S1) and a steady rise in TITR (from 81% to 90%) were observed (Fig. 1h; Supplementary Fig. S7ce and Table S1). After week 32, the patients TITR had readily reached 99% and was maintained thereafter (Fig. 1h, i; Supplementary Table S1), while MAGE, the gold standard of blood glucose variability, was reduced from 5.50mM to 1.60mM (Supplementary Figs. S6d, S7hl and Table S1). Importantly, no episodes of hypoglycemia or severe hyperglycemia were observed during the whole follow-up period of 116 weeks post surgery (Supplementary Fig. S7 and Table S1). Additionally, MMTT revealed a trend of stabilization in glycemic variability after surgery, as manifested by the stable fasting glucose concentrations and the significant reductions in the post-meal glucose concentrations (maximum of 21.3mM at baseline vs maximum of 9.1mM at week 105) (Fig. 1j; Supplementary Table S5). Consistently, the area under the curve (AUC) derived from the 5-point intravenous glucose values decreased to 40% of baseline (Supplementary Fig. S6b), confirmed by the AUCs of the values acquired from CGM (Supplementary Fig. S6c). The hemoglobin A1c levels decreased from 6.6% (baseline) to 5.5% (week 85) and 4.6% (week 113) (Fig. 1h; Supplementary Table S1).

Notably, the insulin requirements were reduced gradually until complete withdrawal at the end of week 11 (Fig. 1h), and the oral antidiabetic medications were tapered since week 44 and discontinued at weeks 48 (acarbose) and 56 (metformin) (Supplementary Fig. S6a).

The average post-surgery fasting C-peptide level (0.68 nmol/L) increased by 3-fold when compared to pre-surgery level (Fig. 1k; Supplementary Table. S5). Notably, the secretions of C-peptide (Fig. 1k) and insulin (Fig. 1l) measured by MMTT revealed significant elevations compared to those of the pre-surgery tests, as confirmed by the AUCs (Supplementary Fig. S6b).

Collectively, we report the first-in-human tissue replacement therapy using autologous E-islets for a T2D patient with impaired islet function. The first 27-month data revealed significant improvements in glycemic control, and provided the first evidence that stem cell-derived islet tissues can rescue islet function in late-stage T2D patients. The grafts were well tolerated with no tumor formation or severe graft-related adverse events.

The precedent clinical trials using cadaveric islets or encapsulated hPSC-derived PPs10,11, along with our study, have provided encouraging evidence that islet tissue replacement is an effective cure for diabetic patients. Notably, the derivation of islet tissues from either hPSCs or EnSCs provides unprecedented new sources for tissue-replacement therapy. Despite the common proof-of-concept purpose, there are some distinctions among the published trials10,11 and ours. First, EnSC-based islet regeneration system is unique, in that EnSCs are nontumorigenic in vivo12 and amenable for efficient mass production of islets as they are endoderm-specific and developmentally closer to pancreatic lineages. Second, our pilot study chose a T2D rather than T1D patient, which not only precluded the interference from autoimmune conditions for the assessment of engraftment and functionality of E-islets but also extended the scope of indications for islet transplantation. As for the limitations of this study, we cannot completely rule out the possibility that the residual endogenous islets benefitted from the surgery and acquired functional improvements. Therefore, an increase in sample size and additional trials of T1D patients with complete loss of islet cells will help draw definitive conclusions on the causative role of E-islets in the achievement of glycemic targets.

Future studies are warranted to address the pharmacodynamics of stem cell-derived islets as a drug, to extend the application of stem cell-derived islet transplantation to other subtypes of diabetes, and to generate universal islets as off-the-shelf products to cure diabetes without the need for immunosuppression.

See the article here:
Treating a type 2 diabetic patient with impaired pancreatic islet function by personalized endoderm stem cell-derived ... - Nature.com

Regenerative Medicine Market Size Expected to Reach USD 154.05 Bn by 2033 – GlobeNewswire

Ottawa, April 26, 2024 (GLOBE NEWSWIRE) -- According to Precedence Research, the global regenerative medicine market size is calculated at USD 35.82 billion in 2024.Regenerative medicines are used in advancing healthcare, such as new treatments for injuries and diseases and gene therapies.

Regenerative Medicine Market Revenue, By Region ($ Million)

Regenerative Medicine Market Revenue, By Product ($ Million)

Immediate Delivery Available | Buy This Premium Research Report@ https://www.precedenceresearch.com/checkout/1176

The regenerative medicine market deals with regenerating, engineering, or replacing animal or human organs, cells, or tissues to restore normal functions. Regenerative medicine can include growing organs or tissues in laboratories and then implanting them into the body when it does not recover itself. Regenerative medicines focus on applying and developing new treatments to restore functions and to heal organs or tissues lost because of defects, damages, aging, or diseases.

Regenerative medicines are also used in tissue engineering, cell therapies, stem cells, and immunomodulation therapy. Regenerative medicine helps to support the body in restoring, repairing, and regenerating itself to a state of well-being and for unsatisfied patient needs among many medical specialties.

The Full Study is Readily Available | Download the Sample Pages of this Report@ https://www.precedenceresearch.com/sample/1176

Key Insights

Regional Stance

North America dominated the regenerative medicine market in 2023. The increasing incidence of joint diseases and bone diseases and advanced technologies in stem cell therapy and tissue engineering are the main factors that are causing the growth of the North American region. The emergency requirement for CAR-T cell therapies and rising investments in developing new regenerative drugs in the United States of the North American region help the growth of the market.

The U.S. regenerative medicine market size reached USD 13.44 billion in 2023 and is projected to surpass around USD 83.26 billion by 2033, expanding at a CAGR of 20% from 2024 to 2033.

Rising genetic diseases and trauma incidences contribute to the growth of the market. Stem cell therapy is used in the United States for medical treatments like the treatment of blood disorders and cancer, as well as specific diseases affecting cartilage, skin, and bone. The USFDA regulates the use of stem cell therapy for some stem cell treatments.

Asia-Pacific is estimated to be the fastest-growing region during the forecast period of 2024-2033.The supply chain of regenerative medicine therapies is growing in the Asia-Pacific region. In the CRO/CDMO space of Asia, growth is increasing. They build a hospital network that supports the development of the regenerative medicine market. Countries like Japan, China, India, and South Korea contribute to the growth of the region. Among all the Asian countries, Japan has a very large influence on the biotechnology and pharmaceutical markets, both of which are responsible for the markets growth. China, being the largest economy, provides endless opportunities for the expansion of the market.

You can place an order or ask any questions, please feel free to contact at sales@precedenceresearch.com | +1 650 460 3308

Scope of Regenerative Medicine Market

Report Highlights

By Product Type

The tissue engineering segment dominated the regenerative medicine market in 2023. Tissue engineering combined with inductive factors, cells, and scaffolds to replace or regenerate diseased or damaged tissue. Regenerative medicine is used in tissue engineering, along with other procedures, such as immunomodulation, cell-based therapy, and gene therapy, to induce organ or tissue regeneration in vivo. Regenerative medicine is a large field that includes tissue engineering and self-healing research, where the body uses its system, sometimes with the help of biological materials, to rebuild organs and tissues or to regenerate cells.

The stem cell therapies segment is the fastest-growing during the forecast period.Stem cell therapy is also called regenerative medicine. It helps in repairing injured, dysfunctional, and diseased tissue by the use of stem cells and their derivatives. Applications of cell therapies in regenerative medicines include regenerating damaged cartilage in joints, improving weakened immune systems, urinary problems, neurological disorders, autoimmune diseases, spinal cord injury, and treating cancers, which helps in the growth of the segment.

Personalized your customization here: https://www.precedenceresearch.com/customization/1176

By Application Type

The musculoskeletal application segment dominated the regenerative medicine market in 2023. Treatment and diagnosis of problems arising from the musculoskeletal systems are treated with the help of regenerative medicine. This includes diseases and injuries affecting the bones, joints of spines, limbs, and muscles. PRP, MSC therapy, and IRAP-induced ACS are regenerative medicines that are used for musculoskeletal injuries.

The oncology application segment is the fastest-growing during the forecast period.Oncology, also known as cancer immunotherapy, is a form of regenerative medicine. Cancer immunotherapy helps in engineering, replacing, regenerating, or activating the immune system to fight against cancer. Cancer immunotherapy is the most commonly used and promoted form of regenerative medicine. Regenerative medicine is used to help cancer patients understand cancer biology and cancer treatment. MSC regenerative medicine may used for the treatment of tumor sites.

Market Dynamics

Driver

Rising senior population

An increased proportion of the senior population is the main driver of the regenerative medicine market. As the senior population increases, degenerative problems and age-related illnesses increase, and this leads to the need for treatments and contributes to the growth of the market. For the treatments of these patients, regenerative medicine is required as age increases. There is a slight decrease in the regenerative properties of many tissues due to age-dependent changes in tissues. Regenerative medicines and anti-aging are involved in the process of reducing harmful elements that cause death or diseases in cells and also modifying these cells to get back to their healthy, normal functions.

Restraint

Reimbursement and manufacturing

The significant challenge for regenerative medicine nowadays is reimbursement and manufacturing, both of which are barriers to the regenerative medicine market. In the fast-moving field of gene therapy and cell therapy, regulation and policy may fail to keep up with scientific innovation. In the manufacturing case, developers and regulators struggle to standardize processes essential for the transition from the making of small-scale batches of therapies to use in clinical trials to the high-scale batches essential by full-scale marketing. The manufacturing processes of gene therapies and cell therapies are more complex than those of other drugs.

Browse More Insights:

AI in Pharmaceutical Market: The global AI in pharmaceutical market size was valued at USD 908 million in 2022 and is expected to reach over USD 11813.56 million by 2032, poised to grow at a compound annual growth rate (CAGR) of 29.30% from 2023 to 2032.

Drug Discovery Services Market: The global drug discovery services market size was valued at USD 21 billion in 2022 and is expected to hit USD 69.72 billion by 2032, poised to grow at a compound annual growth rate (CAGR) of 12.80% from 2023 to 2032.

Small Molecule API Market: The global small molecule API market size was valued at USD 184.8 billion in the year 2023and is expected to hit around USD 299.2 billion by 2032 with an increasing CAGR of 5.5% during the forecast period 2023 to 2032.

Drug Discovery Market: The global drug discovery market size was valued at US$ 55.46 billion in 2022 and is expected to be worth around US$ 133.11 billion by 2032, growing at a CAGR of 9.2% from 2023 to 2032.

Small Molecule Drug Discovery Market: The global small molecule drug discovery market size was exhibited at USD 75.96 billion in 2022 and is projected to hit around USD 163.76 billion by 2032, growing at a CAGR of 7.97% during the forecast period 2023 to 2032.

Small Molecule Immunomodulators Market: The global small molecule immunomodulators market size reached USD 156.2 billion in 2022 and is projected to hit around USD 283.70 billion by 2032, registering a CAGR of 6.20% during the forecast period from 2023 to 2032.

Artificial Intelligence (AI) In Drug Discovery Market: The global artificial intelligence (AI) in drug discovery market size was estimated at USD 1.4 billion in 2022 and is projected to hit around USD 9.7 billion by 2032, registering growth at a CAGR of 21.36% from 2023 to 2032.

Opportunity

Increase in tissue engineering.

Tissue engineering become a transformative field in modern healthcare, providing a creative approach toward replacing, regenerating, and repairing lost or damaged organs or tissues. This helps address the limitations of traditional therapies, revolutionize medical treatments, and improve the quality of life for many individuals. As technological capabilities and scientific understanding increase, tissue engineering development will help and contribute to the growth of the regenerative medicine market. Tissue engineering helps in a combination of biochemical biomaterials and cell factors for creating functional tissues, which are copies of natural tissue functions and properties. At that time, regenerative medicine helps in natural healing mechanisms to replace damaged organs or tissues. These also help to offer personalized therapeutic solutions and overcome the challenges of reducing chronic diseases and organ shortages for transplantation.

Recent Developments

Regenerative Medicine Market Key Players

Market Segmentation

By Product

By Material

By Application

By End User

By Geography

Immediate Delivery Available | Buy This Premium Research Report@ https://www.precedenceresearch.com/checkout/1176

You can place an order or ask any questions, please feel free to contact at sales@precedenceresearch.com | +1 650 460 3308

Unlocking Market Insights through Data Excellence

The "Precedence Statistics" flexible dashboard is a powerful tool that offers real-time news updates, economic and market forecasts, and customizable reports. It can be configured to support a wide range of analysis styles and strategic planning needs. This tool empowers users to stay informed and make data-driven decisions in various scenarios, making it a valuable asset for businesses and professionals looking to stay ahead in today's dynamic and data-driven world.

To Access our Premium Real-Time Data Intelligence Tool, Visit: http://www.precedencestatistics.com

About Us

Precedence Research is a worldwide market research and consulting organization. We give an unmatched nature of offering to our customers present all around the globe across industry verticals. Precedence Research has expertise in giving deep-dive market insight along with market intelligence to our customers spread crosswise over various undertakings. We are obliged to serve our different client base present over the enterprises of medicinal services, healthcare, innovation, next-gen technologies, semi-conductors, chemicals, automotive, and aerospace & defense, among different ventures present globally.

Web: https://www.precedenceresearch.com

Our Blogs:

https://www.towardshealthcare.com

https://www.towardspackaging.com

https://towardsautomotive.com

For Latest Update Follow Us:

Linkedin | Facebook | Twitter

Go here to see the original:
Regenerative Medicine Market Size Expected to Reach USD 154.05 Bn by 2033 - GlobeNewswire

John Cleese says he’s been spending 17,000 annually on stem cell therapy to ‘buy a few extra years’ – Yahoo News UK

John Cleese has revealed he has been spending 17,000 every 12 months for the past 20 years on stem cell therapy in an attempt to combat the effects of ageing.

The Monty Python star and co-creator, who also opened up about being surprisingly poor despite his five-decade career, said he doesnt look bad for his age after getting stem cells from Switzerland.

Stem cells can act as a repair system for the body and are sometimes used in regenerative therapies for long-term conditions like Crohns disease. The potential benefits of stem cells as an anti-ageing remedy include cell rejuvenation, reduced risk of age-related diseases and improved organ function.

In an interview with Saga Magazine, the comedian said: These cells travel around the body and when they discover a place that needs repair, theyll then change into the cells that you want for repair, so they might become cartilage cells or liver cells.

So I think thats why I dont look bad for 84.

He admitted he spends approximately 17,000 every 12 to 18 months.

But if youre buying yourself a few extra years, I think its worth it, he said.

Cleese also said his wife, jewellery designer Jennifer Wade, 52, keeps me young.

Addressing their 32-year age gap, he said: A lot of people comment and then the moment they actually see us together for two minutes they say oh, I get it and it never arises again.

What I love is that shes 30 years younger than I am, but she keeps me young.

I mean, it is sad to think I shall die some time before she will, but Im in pretty good health.

Im not fit, but the way I put it is the doctors dont yet know what Im going to die of.

In the same interview, Cleese said he is surprisingly poor after his five-decade career.

He joked that his $20m divorce from his ex-wife Alyce Faye Eichelberger resulted in him having to work in his eighties.

Cleese was married to Eichelberger, an American psychotherapist, for 16 years before their split in 2008. Eichelberger received a $20m (16m) divorce settlement, after which Cleese travelled the world with The Alimony Tour to raise the money.

When the interviewer notes Cleeses continued busy work schedule despite his elevated age, he refers to Eichelberger in his response: Well, youre quite right, but theres only one person responsible for that.

He continues: Can you believe when I met her, I had a beautiful house in Holland Park and no mortgage and when I broke up with her, I had a flat in Sloane Square and a full mortgage? How they figured out she was worth $20m, I have no idea.

The interview also mentions that Cleese does not own a car or property, having handed over the short lease of his flat to his current wife, Wade.

Although he states that this decision made a huge difference to Wade, Cleese said: But Im surprisingly poor.

I never thought it was necessary to own a great deal. The most important thing is to have enough money to have some really good food, buy clothes twice a year and have nice holidays.

The May 2024 edition of Saga Magazine will be released next week.

The rest is here:
John Cleese says he's been spending 17,000 annually on stem cell therapy to 'buy a few extra years' - Yahoo News UK

Stem cell drive to take place May 26th – CKPGToday.ca

Were trying to increase the diversity of the donor registry in order to help mitigate that and help people find matches whenever they are in need of a transplant for their treatment. Jayda Third

Crime

Prince George RCMP look for suspect in gas station robbery with unique tattoo

stem cell drive

Stem cell drive to take place May 26th

EOC funding

Prince George Emergency Operation Centre in need of a permanent facility says city official

The process for donating is simple, you will be asked a few questions to make sure you are eligible, fill out a virtual questionnaire, and then a buccal swab is conducted. A buccal swab is conducted on the inside of your cheek. Once stem cell samples are collected, they are sent away to be analyzed, since we do not have a blood services centre in Prince George, with the last one closing in 2015.

The stem cell drive will take place on Sunday, May 26 from 11am to 4pm in the Agora at UNBC.

X: @AdamBerls

Email: Adam.Berls@pattisonmedia.com

Follow this link:
Stem cell drive to take place May 26th - CKPGToday.ca

Enhancing Fertility: A Case Report of the Frozen Platelet-Rich Plasma Therapy for Thin Endometrium and Poor Ovarian … – Cureus

Specialty

Please choose I'm not a medical professional. Allergy and Immunology Anatomy Anesthesiology Cardiac/Thoracic/Vascular Surgery Cardiology Critical Care Dentistry Dermatology Diabetes and Endocrinology Emergency Medicine Epidemiology and Public Health Family Medicine Forensic Medicine Gastroenterology General Practice Genetics Geriatrics Health Policy Hematology HIV/AIDS Hospital-based Medicine I'm not a medical professional. Infectious Disease Integrative/Complementary Medicine Internal Medicine Internal Medicine-Pediatrics Medical Education and Simulation Medical Physics Medical Student Nephrology Neurological Surgery Neurology Nuclear Medicine Nutrition Obstetrics and Gynecology Occupational Health Oncology Ophthalmology Optometry Oral Medicine Orthopaedics Osteopathic Medicine Otolaryngology Pain Management Palliative Care Pathology Pediatrics Pediatric Surgery Physical Medicine and Rehabilitation Plastic Surgery Podiatry Preventive Medicine Psychiatry Psychology Pulmonology Radiation Oncology Radiology Rheumatology Substance Use and Addiction Surgery Therapeutics Trauma Urology Miscellaneous

Read more here:
Enhancing Fertility: A Case Report of the Frozen Platelet-Rich Plasma Therapy for Thin Endometrium and Poor Ovarian ... - Cureus

Cabinet approves two bills on regenerative medicine –

By Shelley Shan / Staff reporter

The Executive Yuan yesterday approved two bills to govern regenerative medicine that aim to boost development of the field.

Taiwan would reach an important milestone in regenerative medicine development with passage of the regenerative medicine act and the regenerative medicine preparations ordinance, which would allow studies to proceed and treatments to be developed, Deputy Minister of Health and Welfare Victor Wang () told reporters at a news conference after a Cabinet meeting.

Regenerative treatments have been used for several conditions, including cancer by regenerating blood cells and restoring joint function in soft tissue, Wang said.

The draft legislation requires regenerative treatments to undergo human trials, he said.

However, human trials might be waived if patients lives are in danger or in other extenuating circumstances, he added.

Ministry of Health and Welfare Department of Civil Ethics Director-General Liu Yueh-ping () said that waivers for human trials would only be granted when people seek new treatments after other methods have failed.

The draft regenerative medicine bill stipulates that only government-certified medical institutions can administer such treatments, with individuals or organizations that administer them without permission to face fines of NT$2 million to NT$20 million (US$61,391 to US$613,911).

Non-medical institutions would face fines of NT$2 million to NT$20 million for advertising for the use of regenerative medical treatments, the bill says.

The bill also lists the conditions under which medical institutions could proceed with regenerative medical treatments without first conducting human trials.

People must have a life-threatening or severely debilitating disease, and Taiwan cannot have medication, equipment or technology that meets the need, while medical institutions can offer regenerative medical treatments that have been approved by the ministry before the bill is promulgated, it says.

The bill would also upgrade the status of the Ethical Guidelines Governing the Research of Human Embryos and Embryonic Stem Cell Research to law. The guidelines state that embryos and embryonic stem cells used in regenerative medical studies must not be produced through artificial insemination.

Moreover, embryos or embryonic stem cells acquired for research cannot be used to implant nucleated human ova in cell nuclei of other species, the guidelines say.

Researchers would be banned from acquiring research-use embryos for embryonic stem cells or using them to produce chimeric species that have human germ cells, the guidelines say.

The regenerative medicine preparations ordinance covers treatments that contain genes, cells and their derivatives for human use, including gene therapy, cell therapy and tissue engineering.

Pharmaceutical firms seeking to manufacture or import regenerative treatments must have their facilities inspected and registered, and they must be approved and issued a valid license to manufacture or import the products, the bill says.

Licenses would be valid for five years and can be extended for another five years if an extension is applied for three to six months in advanced and it is approved, the bill says.

To facilitate compassionate treatment for people with life-threatening illnesses for which other treatments are not available, the bill authorizes the ministry to approve under certain conditions the use of regenerative products after a phase 2 clinical trial has been completed and the product has been shown to be safe and effective.

Licenses for such products would be valid for five years and cannot be extended.

Both bills have regulations to protect people seeking regenerative treatment or medication.

The ministry has the authority to stop or terminate all or part of a regenerative treatments if a medical provider is facing an unusually high number of liability cases.

Comments will be moderated. Keep comments relevant to the article. Remarks containing abusive and obscene language, personal attacks of any kind or promotion will be removed and the user banned. Final decision will be at the discretion of the Taipei Times.

Go here to read the rest:
Cabinet approves two bills on regenerative medicine -

Induced pluripotent stem cells (iPSCs): molecular mechanisms of induction and applications | Signal Transduction and … – Nature.com

Rowe, R. G. & Daley, G. Q. Induced pluripotent stem cells in disease modelling and drug discovery. Nat. Rev. Genet. 20, 377388 (2019).

Article CAS PubMed PubMed Central Google Scholar

Shi, Y., Inoue, H., Wu, J. C. & Yamanaka, S. Induced pluripotent stem cell technology: a decade of progress. Nat. Rev. Drug Discov. 16, 115130 (2017).

Article CAS PubMed Google Scholar

Takahashi, K. et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131, 861872 (2007).

Article CAS PubMed Google Scholar

Takahashi, K. & Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663676 (2006).

Article CAS PubMed Google Scholar

Yu, J. et al. Induced pluripotent stem cell lines derived from human somatic cells. Science 318, 19171920 (2007).

Article CAS PubMed Google Scholar

Takahashi, K. & Yamanaka, S. A decade of transcription factor-mediated reprogramming to pluripotency. Nat. Rev. Mol. Cell Biol. 17, 183193 (2016).

Article CAS PubMed Google Scholar

Breschi, A., Gingeras, T. R. & Guigo, R. Comparative transcriptomics in human and mouse. Nat. Rev. Genet. 18, 425440 (2017).

Article CAS PubMed PubMed Central Google Scholar

Gharib, W. H. & Robinson-Rechavi, M. When orthologs diverge between human and mouse. Brief. Bioinform. 12, 436441 (2011).

Article PubMed PubMed Central Google Scholar

Lynch, V. J. Use with caution: developmental systems divergence and potential pitfalls of animal models. Yale J. Biol. Med. 82, 5366 (2009).

CAS PubMed PubMed Central Google Scholar

Takebe, T. & Wells, J. M. Organoids by design. Science 364, 956959 (2019).

Article CAS PubMed PubMed Central Google Scholar

Yamanaka, S. Pluripotent Stem Cell-based Cell Therapy- Promise And Challenges. Cell Stem Cell 27, 523531 (2020).

Article CAS PubMed Google Scholar

Gurdon, J. B. The generation of diversity and pattern in animal development. Cell 68, 185199 (1992).

Article CAS PubMed Google Scholar

Kiefer, J. C. Epigenetics in development. Dev. Dyn. 236, 11441156 (2007).

Article CAS PubMed Google Scholar

Tompkins, J. D. Discovering DNA methylation, the history and future of the writing on DNA. J. Hist. Biol. 55, 865887 (2022).

PubMed PubMed Central Google Scholar

Roe, S. A. Matter, life, and generation: eighteen-century embryology and the Haller-Wolff Debate, (Cambridge University Press, 1981).

Kilgour, F. G. William Harvey and his contributions. Circulation 23, 286296 (1961).

Article CAS PubMed Google Scholar

Aulie, R. P. Caspar Friedrich Wolff and his Theoria generationis, 1759. J. Hist. Med. Allied Sci. 16, 124144 (1961).

Article CAS PubMed Google Scholar

Weismann, A. Das Keimplasma; eine Theorie der Vererbung, (Jena, Fischer, 1892).

Waddington, C. H. The Strategy of the Genes; A Discussion of Some Aspects of Theoretical Biology, (Cambridge: Cambridge University Press, 1957).

Nanney, D. L. Epigenetic control systems. Proc. Natl. Acad. Sci. USA 44, 712717 (1958).

Article CAS PubMed PubMed Central Google Scholar

Gurdon, J. B. The developmental capacity of nuclei taken from intestinal epithelium cells of feeding tadpoles. J. Embryol. Exp. Morphol. 10, 622640 (1962).

CAS PubMed Google Scholar

Gurdon, J. B. The transplantation of nuclei between two species of Xenopus. Dev. Biol. 5, 6883 (1962).

Article CAS PubMed Google Scholar

Gurdon, J. B. Adult frogs derived from the nuclei of single somatic cells. Dev. Biol. 4, 256273 (1962).

Article CAS PubMed Google Scholar

Gurdon, J. B. Multiple genetically identical frogs. J. Hered. 53, 59 (1962).

Article CAS PubMed Google Scholar

Gurdon, J. B., Elsdale, T. R. & Fischberg, M. Sexually mature individuals of Xenopus laevis from the transplantation of single somatic nuclei. Nature 182, 6465 (1958).

Article CAS PubMed Google Scholar

Jeltsch, A. & Jurkowska, R. Z. New concepts in DNA methylation. Trends Biochem. Sci. 39, 310318 (2014).

Article CAS PubMed Google Scholar

Riggs, A. D. X inactivation, differentiation, and DNA methylation. Cytogenet. Cell Genet. 14, 925 (1975).

Article CAS PubMed Google Scholar

Robertson, K. D. & Wolffe, A. P. DNA methylation in health and disease. Nat. Rev. Genet. 1, 1119 (2000).

Article CAS PubMed Google Scholar

Schubeler, D. Function and information content of DNA methylation. Nature 517, 321326 (2015).

Article CAS PubMed Google Scholar

Evans, M. J. & Kaufman, M. H. Establishment in culture of pluripotential cells from mouse embryos. Nature 292, 154156 (1981).

Article CAS PubMed Google Scholar

Martin, G. R. Isolation of a pluripotent cell line from early mouse embryos cultured in medium conditioned by teratocarcinoma stem cells. Proc. Natl. Acad. Sci. USA 78, 76347638 (1981).

Article CAS PubMed PubMed Central Google Scholar

Thomson, J. A. et al. Embryonic stem cell lines derived from human blastocysts. Science 282, 11451147 (1998).

Article CAS PubMed Google Scholar

Tada, M., Takahama, Y., Abe, K., Nakatsuji, N. & Tada, T. Nuclear reprogramming of somatic cells by in vitro hybridization with ES cells. Curr. Biol. 11, 15531558 (2001).

Article CAS PubMed Google Scholar

Cowan, C. A., Atienza, J., Melton, D. A. & Eggan, K. Nuclear reprogramming of somatic cells after fusion with human embryonic stem cells. Science 309, 13691373 (2005).

Article CAS PubMed Google Scholar

Davis, R. L., Weintraub, H. & Lassar, A. B. Expression of a single transfected cDNA converts fibroblasts to myoblasts. Cell 51, 9871000 (1987).

Article CAS PubMed Google Scholar

Halder, G., Callaerts, P. & Gehring, W. J. Induction of ectopic eyes by targeted expression of the eyeless gene in drosophila. Science 267, 17881792 (1995).

Article CAS PubMed Google Scholar

Kulessa, H., Frampton, J. & Graf, T. Gata-1 reprograms Avian Myelomonocytic cell-lines into Eosinophils, Thromboblasts, and Erythroblasts. Gene Dev. 9, 12501262 (1995).

Article CAS PubMed Google Scholar

Xie, H., Ye, M., Feng, R. & Graf, T. Stepwise reprogramming of B cells into macrophages. Cell 117, 663676 (2004).

Article CAS PubMed Google Scholar

Wernig, M. et al. In vitro reprogramming of fibroblasts into a pluripotent ES-cell-like state. Nature 448, 318324 (2007).

Article CAS PubMed Google Scholar

Huangfu, D. W. et al. Induction of pluripotent stem cells by defined factors is greatly improved by small-molecule compounds. Nat. Biotechnol. 26, 795797 (2008).

Article CAS PubMed PubMed Central Google Scholar

Huangfu, D. et al. Induction of pluripotent stem cells from primary human fibroblasts with only Oct4 and Sox2. Nat. Biotechnol. 26, 12691275 (2008).

Article CAS PubMed Google Scholar

Hou, P. et al. Pluripotent stem cells induced from mouse somatic cells by small-molecule compounds. Science 341, 651654 (2013).

Article CAS PubMed Google Scholar

Zhu, J. et al. Genome-wide chromatin state transitions associated with developmental and environmental cues. Cell 152, 642654 (2013).

Article CAS PubMed PubMed Central Google Scholar

Apostolou, E. & Hochedlinger, K. Chromatin dynamics during cellular reprogramming. Nature 502, 462471 (2013).

Article CAS PubMed PubMed Central Google Scholar

Apostolou, E. & Stadtfeld, M. Cellular trajectories and molecular mechanisms of iPSC reprogramming. Curr. Opin. Genet. Dev. 52, 7785 (2018).

Article CAS PubMed PubMed Central Google Scholar

Cacchiarelli, D. et al. Integrative analyses of human reprogramming reveal dynamic nature of induced pluripotency. Cell 162, 412424 (2015).

Article CAS PubMed PubMed Central Google Scholar

Nefzger, C. M. et al. Cell type of origin dictates the route to pluripotency. Cell Rep. 21, 26492660 (2017).

Article CAS PubMed Google Scholar

Borkent, M. et al. A serial shRNA screen for roadblocks to reprogramming identifies the protein modifier SUMO2. Stem Cell Rep. 6, 704716 (2016).

Article CAS Google Scholar

Buckley, S. M. et al. Regulation of Pluripotency and cellular reprogramming by the ubiquitin-proteasome system. Cell Stem Cell 11, 783798 (2012).

Article CAS PubMed PubMed Central Google Scholar

Qin, H. et al. Systematic identification of barriers to human iPSC generation. Cell 158, 449461 (2014).

Article CAS PubMed PubMed Central Google Scholar

Simic, M. S. et al. Transient activation of the UPR(ER) is an essential step in the acquisition of pluripotency during reprogramming. Sci. Adv. 5, eaaw0025 (2019).

Article CAS PubMed PubMed Central Google Scholar

Wu, Y. et al. Phospholipid remodeling is critical for stem cell pluripotency by facilitating mesenchymal-to-epithelial transition. Sci. Adv. 5, eaax7525 (2019).

Go here to see the original:
Induced pluripotent stem cells (iPSCs): molecular mechanisms of induction and applications | Signal Transduction and ... - Nature.com

Mechanical stimulation of induced pluripotent stem derived cardiac fibroblasts | Scientific Reports – Nature.com

CFs are the main contributors of cardiac fibrosis development3. The availability of human CFs is limited hampering the field to move forward. To date, CFs can be generated from iPSCs, which could provide an unlimited source of human CFs13,14,15. However, the behaviour of iPSC-CFs in relation to mechanical stimulation had not been investigated yet.

In this study we demonstrated that iPSC-CFs are comparable to primary CFs with regard to the expression of key CF markers at gene and protein levels. Expression of the cardiac markers GATA4 and TCF21 indicate the cardiac lineage of the cells. Furthermore, expression of the mesenchymal markers VIM and PDGFRA as well as the ECM component COL1A1 and the collagen binding receptor DDR2 support their fibroblast phenotype. In addition, we showed that iPSC-CFs respond to pro-fibrotic and mechanical stimulation. TGF- induces CFs transdifferentiation into myofibroblasts and promotes ECM remodelling. Mechanical stimulation in the form of cyclic stretch at physiological levels reduces collagen expression in iPSC-CFs. Interestingly, cyclic stretch also protects against TGF- stimulation, preventing the cells from transdifferentiating into myofibroblasts.

One can only use iPSC-derived cells when they accurately represent their primary counterparts. Key characteristics of CFs are a defined mRNA profile, responsiveness to pro-fibrotic cytokines, interaction with the ECM and mechanical sensitivity. iPSC-CFs generated using the protocol developed by Zhang et al. showed a comparable RNA sequencing profile in iPSC-CFs and primary CFs13. Using our iPSCs lines, following the same protocol we generated iPSC-CFs with an mRNA profile comparable to primary CFs. Furthermore, at a functional level we demonstrated that iPSC-CFs interact with their environment in a similar way as primary CFs, and respond to pro-fibrotic stimulation. These results indicate that iPSC-CFs possess several key characteristics of primary CFs and may be suitable to investigate the behaviour of CFs and develop disease models of cardiac fibrosis.

In order to investigate the behaviour of CFs in their native environment, we next investigated the behaviour of iPSC-CFs under physiologically relevant conditions. In an effort to mimic the dynamic environment of the continuously beating heart, we investigated the effects of cyclic mechanical stretch on iPSC-CFs. The importance of mechanical stimulation has been acknowledged, but the effects of mechanical stimulation on CFs remain controversial in in vitro studies23. On one hand, it has been reported that cyclic stretch may induce transdifferentiation of CFs into myofibroblasts. On the other hand, it has been shown that cyclic stretch may have a protective effect instead. One of the main factors influencing this controversy is the usage of cell sources from different species.

As primary human CFs are limited in availability, iPSC-CFs could provide a representative and stable source of cells to move forward. In order to study how iPSC-CFs and primary CFs behave in a mechanically dynamic environment similar to the heart, cells were exposed to 10% cyclic stretch at 1Hz for 72h19. With this approach, we demonstrated that: Cyclic stretch alone inhibits expression of collagen 1 but does not affect iPSC-CFs transdifferentiation or expression of matrix remodelling genes. In addition, cyclic stretch is protective against TGF- mediated myofibroblast transdifferentiation in iPSC-CFs, resulting in normalised expression of collagen 1, -SMA and matrix remodelling genes such as TIMP1 and MMP1.

The cause of the aforementioned controversy in literature regarding either the pro-fibrotic or anti-fibrotic response of CFs to mechanical stimulation is hard to pin-point; experimental conditions vary widely between studies, such as cell origin, the duration of the experiment, the surface coating and the presence of serum. A common trend in all those studies is that there may be a time-dependent response of stretch. It was shown in primary mouse CFs that the response starts with an initial increase in phosphorylation of AKT, a downstream kinase involved in the transduction of mechanical stimuli24,25. At the gene level, it was shown in primary rat CFs that there is an initial increase in fibrotic markers (i.e. ACTA2, TGFB1, CTGF) after 4 h followed by a reduced increase after 24h26. Roche et al. observed a similar effect in primary rat CFs with an apparent reduced increase of COL1A1 gene expression after 48h compared to 24h27. 72h of cyclic stretch was instead shown to inhibit TGF- induced fibroblast activation in primary human CFs16,18. Furthermore, it has been demonstrated that 96h of cyclic stretch can promote or inhibit the response of primary mouse CFs to a broad spectrum of biochemical stimuli, including TGF-, angiotensin II, interleukin-1 and others17. Overall, it appears that longer stimulation results in a gradual decrease of an initial pro-fibrotic response with eventually cells balancing the fibrotic response to the mechanically active environment in order to reach homeostasis. We may hypothesize that the duration of this response curve is dependent on different factors, including the origin and age of the cells, their culture conditions (surface coating, substrate stiffness, or medium supplementation with serum) and the presence of other cell types23. A clear association between mechanosensing and a response of CFs is apparent, but there is a need for a reproducible cell type to better understand this phenomenon.

TGF- signalling is one of the main pathways involved in the activation of CFs and development of cardiac fibrosis28. Exposure of iPSC-CFs to TGF- promotes the expression of fibrotic and myofibroblast markers, such as -SMA. When stretched however, this effect is diminished. How mechanical changes communicate with the TGF- pathway is not well understood. On one hand, mechanical strain has been shown in tissue to release active TGF- from the ECM, which would promote fibroblast activation29. On the other hand, in this in vitro study mechanical strain appears to inhibit fibroblast activation, indicating that there may be other mechanisms at play in this model. It is unknown whether this anti-fibrotic effect is directly caused by interplay between mechanosensitive complexes and the TGF- pathway. Mechanosensitive receptors such as integrins or mechanoresponsive factors such as YAP/TAZ may communicate with the TGF- pathway30,31. Alternatively, cyclic stretch may have an indirect effect, for example through internalization of extracellular receptors, altering the response to ligand stimulation. Regardless, the field of mechanotransduction in CFs remains requires further investigation.

While iPSCs have started a new era of research, the usage of these cells comes with limitations. iPSC-CFs showed many similarities with primary CFs, but the maturity of iPSC-derived cell lineages remains an important topic of contention. Although maturation is clearly defined for some cell types, such as cardiac myocytes, a clear definition lacks for CFs. The heterogeneity and plasticity of this cellular population under physiological conditions makes it difficult to set well defined standards of mature CFs32. iPSC-CFs present with various characteristics of primary cells, but they differ in several aspects as well. For example, Zhang et al. noted an increased proliferation capacity in iPSC-CFs and foetal CFs compared to adult CFs, indicating the iPSC-CFs may be more foetal-like13. This increased proliferation capacity and ability to stay in an inactivated state while in culture increases the applicability of the iPSC-CFs in research, as it has been demonstrated that CFs which have transdifferentiated into myofibroblast will have an altered response to mechanical stimulation33. In addition, little is known about the electrophysiological characteristics of iPSC-CFs and their interaction with other conducting cells such as cardiomyocytes34. Further electrophysiological characterisation should be performed to better understand the behaviour of these cell in the electrical circuit of the heart.

To conclude, in this study we demonstrated that iPSC-derived CFs show similar gene and protein expression as primary CFs. In addition, pro-fibrotic stimulation promoted transdifferentiation of iPSC-CFs into a myofibroblast phenotype. When stimulated with cyclic stretch, this transdifferentiation is inhibited. Together, the mechano- and TGF--responsive characteristics support the use of iPSC-CFs for physiological relevant disease modelling. Future studies could further dive into the mechanisms driving cardiac fibroblast behaviour and cardiac fibrosis.

Read more from the original source:
Mechanical stimulation of induced pluripotent stem derived cardiac fibroblasts | Scientific Reports - Nature.com

Integrative metabolomics-genomics analysis identifies key networks in a stem cell-based model of schizophrenia … – Nature.com

Cell culture

Eight human iPSC lines were employed in this study (Supplementary TableS1). The cells were cultured using Corning Matrigel hESC-Qualified Matrix (Corning, Cat. No. 354277) coated plates with the use of StemMACS iPS Brew XF Medium (Miltenyi Biotec, Cat. No. 130-104-368) or Essential 8 medium (ThermoFisher Scientific, Cat. No. A157001), in antibiotic-free conditions, and maintained at 37C, 5% CO2. iPSCs were passaged every 35 days using either Accutase or 0.5mM phosphate-buffered saline (PBS)/EDTA. Briefly, when passaging the cells with Accutase, cells were firstly washed with DMEM, 1ml of Accutase was added per 6-well and the cells were incubated at 37C for 34min, to ensure proper cell detachment. After the incubation an equal volume of DMEM was added to the well and the cells were collected and centrifuged at 1200rpm for 3min at 4C. For splitting with PBS/EDTA (ThermoFisher Scientific, Cat. No. 15575020), cells were briefly washed with DMEM, 1ml PBS/EDTA was added per 6-well and the cells were incubated until they started to roughly dissociate. The EDTA was aspirated and the cells or the cell pellet (when splitting using Accutase), were resuspended in fresh medium supplemented with 10M ROCK inhibitor, Y-27632, (Miltenyi Biotec, Cat. No. 130-106-538). The next day the medium was changed back to iPSC medium without ROCK inhibitor. All cell lines were thoroughly characterized for their pluripotency (Supplementary Fig.1A, B) and were tested frequently for mycoplasma contamination.

The generation of cortical progenitors and neurons was performed as described before [27, 28] with minor modifications. Briefly, iPSCs from five 6-wells were collected with Accutase and seeded onto an ES-Matrigel coated 12-well. When 100% confluency was reached, StemMACS iPS Brew XF Medium was replaced by neural induction medium (NIM; DMEM/F12 Glutamax, Neurobasal, 100 mM L-Glutamine, 0.5N-2, 0.5B-27+Vitamin A, 50M Non-Essential Amino Acids, 50M 2-mercaptoethanol, 2.5g/ml insulin, 1M dorsomorphine, 10M SB431542). The medium was changed every day until the appearance of a tightly packed neuroepithelial sheet (NES). NES was passaged with 0.5mM EDTA in a ratio of 1:2 or 1:3 to Corning Matrigel Growth Factor Reduced (GFR) Basement Membrane Matrix (GFR-Matrigel; Corning, Cat. No. 354230) coated plates. The next day, the medium was switched to neural maintenance medium (NMM; DMEM/F12 Glutamax, Neurobasal, 100mM L-Glutamine, 0.5N-2, 0.5B-27+Vitamin A, 50M Non-Essential Amino Acids, 50M 2-mercaptoethanol, 2.5g/ml insulin) and was changed every other day. Upon the appearance of rosettes, 20ng/ml FGF2 (Peprotech, Cat. No. 100-18C) were added to the medium for four days. On the fourth day of FGF2 treatment, the cells were split again with 0.5mM EDTA in a ratio of 1:2 to 1:3 onto GFR-Matrigel coated plates. The medium was switched back to NMM and the cortical progenitors were maintained for about 510 days until neurons accumulated outside of the rosettes. At this point, cells were passaged with Accutase, and 50,000 cells/cm were seeded on poly-L-ornithin/Laminin coated plates for further neuronal differentiation. Alternatively, 24million cells/ml were frozen with neural freezing medium. Neurons differentiated further with half medium changes every two to three days. Samples were harvested at day (d) 0, 7, 16, 27, 50, and 100.

For the DFMO treatment, adherent cell cultures were treated daily with 10M DFMO (difluoromethylornithine hydrochloride hydrate; Merck, Cat. No. D193) starting from the first day of differentiation until the collection of cellular pellet and supernatant for mass spectrometry analysis or fixation for subsequent immunocytochemistry (ICC).

Cells were fixed in 4% paraformaldehyde (PFA; Sigma, Cat. No. 158127-500G) in PBS solution for 20min at room temperature (RT). The non-specific binding was blocked with incubation with blocking buffer (3% bovine serum albumin (BSA), 0.2% Triton 100 in PBS) for 1h at RT. The primary antibody (Ab) was diluted in the blocking buffer in the recommended concentration and the Ab solution was applied overnight at 4C. The following primary Abs were used in the following concentrations: AFP 1:400 (Dako, Cat. No. A000829-2), GAD65/67 1:100 (Abcam, Cat. No. AB183999), GFAP 1:400 (Sigma, Cat. No. G3893-.2ML), Ki67-VioR667 1:200 (Miltenyi, Cat. No.130-120-422), MAP2 1:1,000 (SynapticSystems, Cat. No.188006), NEUN 1:500 (Sigma, Cat. No. ABN78), OCT3/4 1:200 (Szabo-Scandic, Cat. No. GTX101497-100), PAX6 1:500 (Invitrogen, Cat. No. 42-6600), S100b 1:750 (Abcam, Cat. No. ab52642), SMA 1:500 (Abcam, Cat. No. ab7817), SOX1 1:200 (R&D Systems, Cat. No. AF3369), SOX2 1:500 (R&D Systems, Cat. No. MAB2018), TAU 1:200 (Cell Signaling Technology, Cat. No. 4019), TUBB3 1:1,000 (BioLegend, Cat. No. 801202 and Abcam, Cat.No. ab52623), vGLUT 1:100 (SynapticSystems, Cat. No. 135311). The secondary Ab was diluted 1:500 in 1.5% BSA, 0,2% Triton 100 in PBS, and the solution was applied for 2h at RT. The secondary Abs used in this study were: donkey anti-rabbit Alexa FluorTM 488 (ThermoFisher Scientific, Cat. No. A-21206), donkey anti-rabbit Alexa FluorTM 546 (ThermoFisher Scientific, Cat. No. A-10040), donkey anti-mouse Alexa FluorTM 594 (ThermoFisher Scientific, Cat. No. A-21203), donkey anti-mouse Alexa FluorTM 647 (ThermoFisher Scientific, Cat. No. A-31571), donkey anti-goat Alexa FluorTM 594 (ThermoFisher Scientific, Cat. No. A-11058), goat anti-chicken Alexa FluorTM 594 (ThermoFisher Scientific, Cat. No. A32759). Finally, the nuclei were counterstained using 4,6-diamidino-2-phenylindole (DAPI; ThermoFisher Scientific, Cat. No. D21490) in PBS in 1:5000 dilution for 5min at RT. The coverslips were mounted using Aqua-Poly/Mount mounting medium (PolySciences, Cat. No. 18606-20).

Fluorescent pictures were acquired with the Zeiss Axio Observer Z1 inverted fluorescent microscope and the Leica DMi8 inverted microscope. The image acquisition was performed under the same exposure and laser intensity settings for each set of analyses. For each sample, ten random fields of view were acquired, with a minimum of 20 z-stacks collected per field to ensure proper signal coverage. Further image processing was carried out using the ImageJ software. For quantitative fluorescence intensity analysis, maximum intensity projection was applied and the mean fluorescence intensity values were calculated after background noise subtraction. These values were then normalized to the DAPI+ nuclear area to account for variations in cell density in the different fields of view.

Total RNA was extracted from cells using TRI Reagent (Merck, Cat. No. T9424), according to the manufacturers instructions. Genomic DNA was removed through treatment with DNase I (Sigma-Aldrich, Cat. No. AMPD1). Subsequently, 1g of purified RNA was reverse transcribed into cDNA using the RevertAid RT Reverse Transcription Kit (ThermoFisher Scientific, Cat. No. K1691), following the manufacturers guidelines. The expression levels of specific target genes at the mRNA level were quantified via reverse transcription quantitative PCR (RT-qPCR) using the 5 HOT FIREPol EvaGreen qPCR Mix Plus (no ROX) (Solis BioDyne, Cat. No. 08-25-00001-10). Samples were analyzed in technical triplicates to ensure data reliability. Non-template controls (NTCs) were included for each primer pair in every assay to monitor for reagent contamination and primer-dimer formation. To confirm the absence of genomic DNA contamination, random RNA samples were evaluated through gel electrophoresis. The RT-qPCR assays were conducted on the CFX Connect Real-Time PCR Detection System (Bio-Rad). Gene expression levels were normalized to the housekeeping gene ACTB. Relative expression changes were calculated employing the Ct method [29]. The list of the primers used for RT-qPCR assays is shown in Table1.

Total RNA was isolated from cells at six time points during the cortical differentiation and was prepared for paired-end mRNA sequencing. RNA extraction was performed using the TRI Reagent (Merck, Cat. No. T9424) according to the manufacturers guidelines. Genomic DNA digest was performed with the use of the TURBO DNA-free Kit (ThermoFisher Scientific, Cat. No. AM2238). For the library preparation, the Illumina TruSeq RNA Library Prep Kit v2 was used (Illumina, Cat. No. RS-122-2001, RS-122-2002). Quality, as well as concentration of RNA were assessed employing the Agilent RNA 6000 Pico kit (Agilent, cat. no. 5067-1513), Nanodrop, the NEBNext Library Quant Kit for Illumina (New England Biolabs, Cat. No. E7630S) and the Qubit RNA Integrity and Quality (IQ) Assay Kit (ThermoFisher Scientific, Cat. No. Q33222). All the kits were used according to the manufacturers guidelines. Paired-end sequencing was performed with the NextSeq 500/550 v2 Kit (150 cycles) (Illumina).

Low-quality ends and adapter sequences were trimmed using the wrapper Trim Galore!. Reads were mapped to the human reference genome (GRCh38) using the open-source software STAR [30]. The raw counts were generated with the Hypergeometric Optimization of Motif EnRichment (HOMER) suite [31]. All the subsequent analysis was performed using R [32]. Differential gene expression analysis was performed using the DESeq2 package [33]. Raw counts were normalized using the median of ratios (variance stabilization transformation; vst) [34]. Heatmaps were generated with the ClustVis [35] tool, using the z-score of the vst transcriptomic data for every gene. Gene ontology (GO) enrichment analysis was performed using the ShinyGO 0.76 online tool [36].

A likelihood ratio test (LRT) was used to identify the differentially expressed genes (DEGs) of SCZ and control (CTRL) across the multiple time points of neuronal differentiation [32]. The LRT compared the full model containing the covariates sex, batch, time point, and disease with a model reducing the covariates sex, batch, and time point. Statistical values were corrected for FDR using the Benjamini-Hochberg method.

Weighted Gene Correlation Network Analysis (WGCNA) allows the generation of modules that include genes that are co-expressed in the same manner. The vst counts were used to build a co-expression network using the WGCNA [37] package in R [32]. The data were corrected for sex and batch effects using the ComBat function that is implemented in the sva package [38]. The topological overlap measure was calculated using the adjacency matrix. The DynamicTree Cut algorithm, implemented in the WGCNA package, was used to identify the different modules. The gray module contains all the genes that were not assigned to any of the other modules. The module eigengene were calculated. Pearsons correlation was used to compare modules to each other and to the traits SCZ and the differentiation time points in the adjacency matrix. The top 25% of genes with the highest module membership (MM) were identified as hub genes.

Functional enrichment analysis was performed with an input gene ID list using the tool g:GOSt from the g:Profiler [39] R package. Statistical significance was computed and the g:SCS-threshold was corrected at p<0.05.

The cells were washed with 1ml sterile 1x PBS for 60s. After the wash, the cells were scraped using 1ml PBS and the suspension was collected and centrifuged at 4000rpm for 5min, at RT. The cell pellets were kept constantly on dry ice and stored at 80C until further processing. The cell supernatant was collected after a 24-h incubation, centrifuged at 4000rpm for 10min, immediately placed on dry ice and stored at 80C. Samples were analyzed using the biocrates MxP Quant 500 (biocrates life sciences AG, Cat. No. 21094.12). Liquid chromatography-tandem mass spectrometry (LC-MS/MS) was employed to analyze small molecules, including analyte classes such as amino acids, biogenic amines, carboxylic acids, and amino acid-related molecules [40]. Lipid species were measured using flow injection analysis tandem mass spectrometry (FIAMS/MS). Small molecules were quantified with external 7-point calibrations and internal standards and lipids were quantified by internal standards [41]. The raw data were processed by applying a modified 80% rule to reduce the false positive measurements [42]. The actual missing values, i.e., the values over the level of detection (LOD) for one time point but not for another time point, were uniformly at random imputed with a non-zero value between LOD/2 and LOD. Missing values within one class(i.e., timepoints and metabolites) were imputed using the arithmetic mean of the class. Batch effects were corrected by centering the data within the groups(i.e., time points) and batches. The performance of the normalization was assessed by plotting the row standard deviations versus the row means and the principal component analysis (PCA). In addition, variancePartition analysis was performed to evaluate the contribution of each individual component of the study design (i.e., time point, batch, and condition), to the measured variation of each metabolite [43].

For metabolite extraction, cell pellets were resuspended in 500L ice-cold methanol. Metabolites from supernatants (50L) were extracted using 450L 8:1 methanol:water. Fully 13C, 15N labeled amino acid standard (Cambridge Isotope Laboratories, Cat. No. MSK-CAA-1) and 6D-gamma hydroxybutyrate (Sigma-Aldrich, Cat. No. 615587) were spiked into samples at the first step of the extraction. After simultaneous proteo-metabolome liquid-liquid extraction [44], protein content was determined from extracted cellular interphases using a Pierce Micro BCA Protein Assay Kit (Thermo Fisher Scientific, Cat. No. 23235). Dried metabolite samples from cell pellets were dissolved in 20L 0.1% formic acid (FA) or 50L 0.1% FA for the analysis from the supernatant samples. The sample (1L) was injected on an Atlantis Premier BEH C18 AX column (1.7m, 2.1150mm, Waters, 186009361) equilibrated at 40C using an Acquity Premier UPLC system (Waters). A gradient was run at a flowrate of 0.4mL/min with mobile phase A (0.1% FA in water) and mobile phase B (0.1% FA in acetonitrile) as follows: 1min at 1% B, to 40% B in 1min, 40% B to 99% B in 0.5min, hold at 99% B for 1.1min, 99% B to 1% B in 0.1min followed by 1.8min of re-equilibration at 1% B. GABA and Glutamate (Glu) were detected using a Xevo-TQ XS Mass spectrometer (Waters) equipped with an electrospray ionization source running in positive mode. The transitions 104>69 (endogenous GABA), 110>73 (labeled GABA), 148>102 (endogenous Glu) and 154>107 (labeled Glu) were used for quantification. The raw files were processed using MS Quan in waters connect (Waters, V1.7.0.7). The data was further analyzed in R and normalized to the protein content.

To analyze time-related cluster dynamics, the non-parametric clustering algorithm of Short Time-series Expression Miner (STEM) was used [45]. STEM is an online tool that assigns genes or metabolites to significant temporal expression profiles. The Maximum Number of Model Profiles and the Maximum Unit Change in Model Profiles between time points were set to 50 and 2, respectively. Data were normalized to d0. Integrated into the STEM tool is a GO enrichment analysis. All annotations (Biological Process (BP), Molecular Function (MF), and Cellular Component (CC)) were selected and applied. Statistical significance was computed and FDR-corrected at p<0.05.

The network establishment was based on the gene expression and metabolite level changes across the five successive time point comparisons, along the cortical differentiation. The connectivity information for the initial network was acquired from the publicly available recon3D stoichiometric model data set (available at https://www.vmh.life/#downloadview, retrieved in September 2020) [46]. Ultimately, 51 metabolites and 1135 genes were matched with their corresponding IDs.

Briefly, the construction of the network was performed based on the following steps. Initially, all the reactions associated with any of the target genes were extracted. The metabolites associated with these reactions were identified and the educt-product stoichiometry was applied for every metabolite involved in the network. Subsequently, the reaction data were filtered to extract and proceed only with the genes and metabolites measured in our dataset. The network was further enriched with protein-protein interaction information, derived using the signor database (available at https://signor.uniroma2.it/downloads.php, retrieved in September 2020) [47]. Finally, the network vertices were constructed after examining the unique metabolites and genes, existing in the edge dataset and were further enriched with vertex attributes, such as the vertex type (i.e., gene/metabolite). Log2 fold changes (log2FC) were converted to a color gradient scale, ranging from blue (indicating a downregulation compared to the previous time point) to red (indicating upregulation).

Extraction of subnetworks from the parental network, was based on assigning membership to the pathways, as defined by the KEGG pathway database, and selecting the subnetwork that included the highest number of differentially expressed genes and metabolites, with the closest degree distribution of the vertices. Pie charts with five equal fractions were used in order to visualize the fold changes occurring across a single metabolite or gene, corresponding to the transitions between two succeeding time points. Additionally, ellipses were used for visualizing the metabolites, while the genes were visualized with circles.

Metabolites that were needed as substantial interconnections between measured metabolites, but were not measured in our dataset, were visualized as small dots. The position for every node was provided as coordinates on a 2D plane. Network visualizations were performed using the R igraph package [48].

Follow this link:
Integrative metabolomics-genomics analysis identifies key networks in a stem cell-based model of schizophrenia ... - Nature.com

Using stem cell-derived heart muscle cells to advance heart regenerative therapy – EurekAlert

image:

Study shows that cardiac spheroids, derived from human induced pluripotent stem cells, can be easily transported and injected into damaged areas of the heart to promote its regeneration and recovery of function.

Credit: Hideki Kobayashi from Shinshu University

Regenerative heart therapies involve transplanting cardiac muscle cells into damaged areas of the heart to recover lost function. However, the risk of arrhythmias following this procedure is reportedly high. In a recent study, researchers from Japan tested a novel approach that involves injecting cardiac spheroids, cultured from human stem cells, directly into damaged ventricles. The highly positive outcomes observed in primate models highlight the potential of this strategy.

Cardiovascular diseases are still among the top causes of death worldwide, and especially prevalent in developed countries. Myocardial infarctions, commonly known as heart attacks, are on the rise, resulting in a significant number of deaths each year.

Heart attacks typically kill millions of cardiac muscle cells, leaving the heart in a weakened state. Since mammals cannot regenerate cardiac muscle cells on their own, heart transplants are currently the only clinically viable option for patients suffering (or likely to suffer) heart failure. Given that full heart transplants are expensive and donors difficult to come by, it is no surprise that alternative therapies are highly sought after by the medical community.

One promising strategy that has been steadily gaining traction is using human induced pluripotent stem cells (HiPSCs) for regenerative heart therapy. Simply put, HiPSCs are cells derived from mature cells that can be effectively reprogrammed into a completely different cell type, such as cardiac muscle cells (cardiomyocytes). By transplanting or injecting cardiomyocytes derived from HiPSCs into damaged areas of the heart, it is possible to recover some lost functionality. Unfortunately, studies have reported that this approach can increase the risk of arrythmias, posing a major hurdle to clinical trials.

In a recent study, a Japanese research team from Shinshu University and Keio University School of Medicine, tested a new strategy for regenerative heart therapy that involves injecting cardiac spheroids derived from HiPSCs into monkeys with myocardial infarction. This study, published on April 26, 2024, in the journal Circulation, was led by Professor Yuji Shiba from the Department of Regenerative Science and Medicine, Shinshu University.

The team included Hideki Kobayashi, the first author, and Koichiro Kuwahara from the Department of Cardiovascular Medicine, Shinshu University School of Medicine, as well as Shugo Tohyama, and Keiichi Fukuda from the Department of Cardiology, Keio University School of Medicine, among others.

In their novel approach, the researchers cultivated HiPSCs in a medium that led to their differentiation into cardiomyocytes. After carefully extracting and purifying cardiac spheroids (three-dimensional clusters of cardiac cells) from the cultures, they injected approximately 6 107 cells into the damaged hearts of crab-eating macaques (Macaca fascicularis). They monitored the condition of the animals for twelve weeks, taking regular measurements of cardiac function. Following this, they analyzed the monkeys hearts at the tissue level to assess whether cardiac spheroids could regenerate the damaged heart muscles.

First, the team verified the correct reprogramming of HiPSCs into cardiomyocytes. They observed, via cellular-level electrical measurements, that the cultured cells exhibited potential patterns typical of ventricular cells. The cells also responded as expected to various known drugs. Most importantly, they found that the cells abundantly expressed adhesive proteins such as connexin 43 and N-cadherin, which would promote their vascular integration into an existing heart.

Afterwards, the cells were transported from the production facility at Keio University to Shinshu University, located 230 km away. The cardiac spheroids, which were preserved at 4 C in standard containers, withstood the four-hour journey without problem. This means that no extreme cryogenic measures would be needed when transporting the cells to clinics, which would make the proposed approach less expensive and easier to adopt.

Finally, the monkeys received injections of either cardiac spheroids or a placebo directly into the damaged heart ventricle. During the observation period, the researchers noted that arrythmias were very uncommon, with only two individuals experiencing transient tachycardia (fast pulse) in the first two weeks among the treatment group. Through echocardiography and computed tomography exams, the team confirmed that the hearts of monkeys that received treatment had better left ventricular ejection after four weeks compared to the control group, indicating a superior blood pumping capability.

Histological analysis ultimately revealed that the cardiac grafts were mature and properly connected to pre-existing existing tissue, cementing the results of previous observations. HiPSC-derived cardiac spheroids could potentially serve as an optimal form of cardiomyocyte products for heart regeneration, given their straightforward generation process and effectiveness, remarks Assistant Professor Kobayashi. We believe that the results of this research will help solve the major issue of ventricular arrhythmia that occurs after cell transplantation and will greatly accelerate the realization of cardiac regenerative therapy, he further adds.

Although tested in monkeys, it is worth noting that the cardiac spheroid production protocol used in this study was designed for clinical application in humans. The favorable results obtained thus far are sufficient to provide a green light for our clinical trial, called the LAPiS trial. We are already employing the same cardiac spheroids on patients with ischemic cardiomyopathy, comments Asst. Prof. Kobayashi.

Let us all hope for a resounding success in the LAPiS trial, paving the way for expanded and effective treatment avenues for people suffering from heart problems.

###

About Shinshu University

Shinshu Universityis a national university founded in 1949 and located nestling under the Japanese Alps in Nagano known for its stunning natural landscapes. Our motto, "Powered by Nature - strengthening our network with society and applying nature to create innovative solutions for a better tomorrow" reflects the mission of fostering promising creative professionals and deepening the collaborative relationship with local communities, which leads to our contribution to regional development by innovation in various fields. Were working on providing solutions for building a sustainable society through interdisciplinary research fields: material science (carbon, fiber and composites), biomedical science (for intractable diseases and preventive medicine) and mountain science, and aiming to boost research and innovation capability through collaborative projects with distinguished researchers from the world. For more information visit https://www.shinshu-u.ac.jp/english/ or follow us on X (Twitter) @ShinshuUni for our latest news.

About Assistant Professor Hideki Kobayashi

Prof. Hideki Kobayashi became a faculty member at Shinshu University Graduate School of Medicine in 2017, focusing his expertise in cardiology and cardiac electrophysiology. With a prolific publication record, he has contributed to over 25 papers in these fields. His professional affiliations include membership in esteemed organizations such as the Japan Society of Internal Medicine, the Japan Circulation Society, the Japanese Heart Rhythm Society and the International Society of Cardiology Research.

About Professor Yuji Shiba

Prof. Yuji Shiba obtained MD and PhD degrees from Shinshu University in 1998 and 2007, respectively, and has remained closely affiliated with the institution throughout his career. Since 2017, he has been a full Professor at the Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research at Shinshu University. His research endeavours primarily focus on cardiac regeneration using stem cells, an emerging biotechnological field. He has published over 45 papers on these topics, as well as over 10 books and book chapters. He is a member of the Japan Society of Internal Medicine and the Japan Circulation Society.

Experimental study

Animals

Regeneration of non-human primate hearts with human induced pluripotent stem cell-derived cardiac spheroids

26-Apr-2024

The authors have no competing interests to declare.

Continue reading here:
Using stem cell-derived heart muscle cells to advance heart regenerative therapy - EurekAlert