Page 41«..1020..40414243..5060..»

Longeveron Inc. Announces U.S. FDA Grants Lomecel-B Regenerative Medicine Advanced Therapy Designation for the Treatment of Mild Alzheimer’s Disease -…

Posted: July 11, 2024 at 2:42 am

Longeveron Inc. announced that the U.S. Food and Drug Administration (FDA) granted Regenerative Medicine Advanced Therapy (RMAT) designation to Lomecel-B for the treatment of mild Alzheimer?s Disease. Lomecel-B is a proprietary, scalable, allogeneic cellular investigational therapy being evaluated across multiple indications, including Alzheimer?s Disease (Phase 2a completed), Aging-related Frailty (Phase 2b completed) and hypoplastic left heart syndrome (HLHS) (Phase 2b on-going). To the Company's knowledge based on publicly available information, Lomecel-B is the first cellular therapeutic candidate to receive RMAT designation for Alzheimer?s Disease.

Longeveron previously announced top-line results for the CLEAR MIND Phase 2a clinical trial on October 5, 2023, and reported additional clinical data and imaging biomarker results on December 20, 2023. Full study results for CLEAR MIND will be reported in a Featured Research Oral Presentation at the 2024 Alzheimer?s Association International Conference (AAIC) on July 28, 2024. Established under the 21st Century Cures Act, RMAT designation is a dedicated program designed to expedite the drug development and review processes for promising pipeline regenerative medicine products, including cell therapies.

A regenerative medicine therapy is eligible for RMAT designation if it is intended to treat, modify, reverse or cure a serious or life-threatening disease or condition, and preliminary clinical evidence indicates that the drug or therapy has the potential to address unmet medical needs for such disease or condition. Similar to Breakthrough Therapy designation, RMAT designation provides the benefits of intensive FDA guidance on efficient drug development, including the ability for early interactions with FDA to discuss surrogate or intermediate endpoints, potential ways to support accelerated approval and satisfy post-approval requirements, and potential priority review of the biologics license application (BLA) if Priority Review designation is granted following BLA submission.

Excerpt from:
Longeveron Inc. Announces U.S. FDA Grants Lomecel-B Regenerative Medicine Advanced Therapy Designation for the Treatment of Mild Alzheimer's Disease -...

Posted in Regenerative Medicine | Comments Off on Longeveron Inc. Announces U.S. FDA Grants Lomecel-B Regenerative Medicine Advanced Therapy Designation for the Treatment of Mild Alzheimer’s Disease -…

Longeveron Announces U.S. FDA Grants Lomecel-B Regenerative Medicine Advanced Therapy (RMAT) Designation for the Treatment of Mild Alzheimers Disease…

Posted: July 11, 2024 at 2:42 am

Longeveron announced that the U.S. FDA granted Regenerative Medicine Advanced Therapy (RMAT) designation to Lomecel-B for treating mild Alzheimers Disease. This makes Lomecel-B the first cellular therapeutic candidate to receive RMAT for Alzheimers. The therapy is being evaluated in multiple conditions, including Alzheimers (Phase 2a completed), Aging-related Frailty (Phase 2b completed), and Hypoplastic Left Heart Syndrome (HLHS) (Phase 2b ongoing). The CLEAR MIND Phase 2a trial indicated that Lomecel-B slowed/prevented disease progression compared to placebo, meeting safety and efficacy endpoints. Full results will be presented in July 2024. RMAT designation aims to expedite the drug development process and offers benefits like FDA guidance and potential priority review.

Positive

Receiving the RMAT designation is a significant advancement in Longeveron's journey, especially since Lomecel-B appears to be the first cellular therapeutic candidate to obtain such recognition for treating Alzheimer's Disease. The RMAT status, akin to the Breakthrough Therapy designation, implies that preliminary clinical data is promising enough to warrant expedited review processes. This designation potentially translates into a faster path to market, provided subsequent trials reinforce the positive outcomes observed in the Phase 2a.

Primarily, the designation underscores early indicators of efficacy. The Phase 2a trial demonstrated the slowing or prevention of disease progression, which is quite notable given the limited therapeutic options currently available for Alzheimer's. If these results hold true in further phases, it could revolutionize treatment paradigms.

For investors, the RMAT designation should be seen as a validation of both the science behind Lomecel-B and the strategic decisions of Longeveron. However, it's essential to remember that while the RMAT can speed up the process, it doesn't guarantee final FDA approval. Therefore, keeping an eye on upcoming clinical results, especially from the CLEAR MIND study, is crucial.

The financial landscape of Longeveron could witness substantial shifts due to the RMAT designation for Lomecel-B. Market sentiment often reacts positively to such regulatory advancements, potentially driving up stock prices in the short term. The RMAT designation also enhances the company's profile, making it more attractive to potential investors and partners who are interested in regenerative medicine and neurodegenerative diseases.

In the long run, successful commercialization of Lomecel-B for Alzheimer's could position Longeveron as a leader in a high-demand yet underserved market. With Alzheimer's Drug sales expected to continue growing due to the aging population, the financial implications of successfully bringing an approved treatment to market could be immense.

However, investors should exercise caution. The road to commercialization is fraught with regulatory and developmental hurdles. While the RMAT designation is a positive step, it's just one part of a longer journey. Upcoming clinical data and FDA interactions will provide more definitive indicators of commercial viability.

While my primary focus is oncology, the implications of Lomecel-B's RMAT designation can be understood from a broader biomedical perspective. The RMAT designation emphasizes the therapy's potential to meet unmet medical needs in Alzheimer's Disease, which is a huge medical challenge with significant overlap in the methodologies used in cancer and regenerative medicine research.

The promising early data showing a slowing of disease progression aligns with what we aim for in oncologydisease modification. From a clinical standpoint, this kind of designation is awarded when early clinical trials show significant promise, which in this case, indicates Lomecel-B might have a substantial impact on disease management.

Given the complexity of Alzheimers and the limited treatment options, any therapy that can demonstrate efficacy in the early stages is worth serious attention. However, it's important for investors to recognize that medical breakthroughs undergo rigorous scrutiny and require consistent, replicable results across larger patient populations before they can be deemed effective and safe for widespread use.

- Lomecel-Bappears to be the first cellular therapeutic candidate to receive RMAT designation for Alzheimers Disease

MIAMI, July 10, 2024 (GLOBE NEWSWIRE) -- Longeveron Inc. (NASDAQ: LGVN), a clinical stage regenerative medicine biotechnology company developing cellular therapies for certain life-threatening and chronic aging-related conditions, today announced that the U.S. Food and Drug Administration (FDA) granted Regenerative Medicine Advanced Therapy (RMAT) designation to Lomecel-Bfor the treatment of mild Alzheimers Disease. Lomecel-Bis a proprietary, scalable, allogeneic cellular investigational therapy being evaluated across multiple indications, including Alzheimers Disease (Phase 2a completed), Aging-related Frailty (Phase 2b completed) and hypoplastic left heart syndrome (HLHS) (Phase 2b on-going). To the Company's knowledge based on publicly available information, Lomecel-Bis the first cellular therapeutic candidate to receive RMAT designation for Alzheimers Disease.

The RMAT designation is an important milestone for Longeveron and the Lomecel-Bprogram that recognizes the potential of our cellular therapy to have a positive impact on patients afflicted with this devastating disease, said Joshua Hare, Co-founder, Chief Science Officer and Chairman of the Board at Longeveron. Alzheimers Disease is a neurodegenerative disorder that leads to progressive memory loss and death and currently has very limited therapeutic options. In the CLEAR MIND Phase 2a clinical trial, Lomecel-Bdemonstrated an overall slowing/prevention of disease worsening compared to placebo. The trial achieved the primary safety and secondary efficacy endpoints and showed statistically significant improvements in pre-specified clinical and biomarker endpoints in specific Lomecel-B groups compared to placebo.

Longeveron previously announced top-line results for the CLEAR MIND Phase 2a clinical trial on October 5, 2023, and reported additional clinical data and imaging biomarker results on December 20, 2023. Full study results for CLEAR MIND will be reported in a Featured Research Oral Presentation at the 2024 Alzheimers Association International Conference (AAIC) on July 28, 2024.

We are thrilled to receive the RMAT designation for Lomecel-Bfor the treatment of mild Alzheimers Disease and we look forward to meeting with the FDA to discuss the path forward and the development plans for Alzheimers Disease in the very near future, said Nataliya Agafonova, M.D., Chief Medical Officer at Longeveron.

The RMAT designation is the fourth special regulatory designation received by Longeveron for Lomecel-B. The Lomecel-BHLHS program has received three separate and distinct FDA designations: Orphan Drug designation, Fast Track designation, and Rare Pediatric Disease designation.

We are happy to receive this important RMAT designation as it continues to validate the work we have done so far and allows us to have the important dialogue with the FDA to advance our work and potentially bring this investigational therapeutic option to the many patients suffering from Alzheimers Disease, said Wael Hashad, Chief Executive Officer and Board Member at Longeveron.

Established under the 21st Century Cures Act, RMAT designation is a dedicated program designed to expedite the drug development and review processes for promising pipeline regenerative medicine products, including cell therapies. A regenerative medicine therapy is eligible for RMAT designation if it is intended to treat, modify, reverse or cure a serious or life-threatening disease or condition, and preliminary clinical evidence indicates that the drug or therapy has the potential to address unmet medical needs for such disease or condition. Similar to Breakthrough Therapy designation, RMAT designation provides the benefits of intensive FDA guidance on efficient drug development, including the ability for early interactions with FDA to discuss surrogate or intermediate endpoints, potential ways to support accelerated approval and satisfy post-approval requirements, and potential priority review of the biologics license application (BLA) if Priority Review designation is granted following BLA submission.

About Longeveron Inc.

Longeveron is a clinical stage biotechnology company developing regenerative medicines to address unmet medical needs. The Companys lead investigational product is Lomecel-B, an allogeneic medicinal signaling cell (MSC) therapy product isolated from the bone marrow of young, healthy adult donors. Lomecel-B has multiple potential mechanisms of action encompassing pro-vascular, pro-regenerative, anti-inflammatory, and tissue repair and healing effects with broad potential applications across a spectrum of disease areas. Longeveron is currently pursuing three pipeline indications: hypoplastic left heart syndrome (HLHS), Alzheimers Disease, and Aging-related Frailty. The Lomecel-BHLHS program has received three separate and distinct FDA designations: Orphan Drug designation, Fast Track designation, and Rare Pediatric Disease designation. For more information, visit http://www.longeveron.com or follow Longeveron on LinkedIn, X, and Instagram.

Forward-Looking Statements

Certain statements in this press release that are not historical facts are forward-looking statements made pursuant to the safe harbor provisions of the Private Securities Litigation Reform Act of 1995, which reflect managements current expectations, assumptions, and estimates of future operations, performance and economic conditions, and involve risks and uncertainties that could cause actual results to differ materially from those anticipated by the statements made herein. Forward-looking statements are generally identifiable by the use of forward-looking terminology such as believe, expects, may, looks to, will, should, plan, intend, on condition, target, see, potential, estimates, preliminary, or anticipates or the negative thereof or comparable terminology, or by discussion of strategy or goals or other future events, circumstances, or effects and include, but are not limited to, the potential for Lomecel-Bto be an effective treatment for Alzheimers Disease or achieve U.S. FDA approval as a Regenerative Medicine Advanced Therapy (RMAT). Factors that could cause actual results to differ materially from those expressed or implied in any forward-looking statements in this release include, but are not limited to, market and other conditions, our limited operating history and lack of products approved for commercial sale; adverse global conditions, including macroeconomic uncertainty; inability to raise additional capital necessary to continue as a going concern; our history of losses and inability to achieve profitability going forward; the absence of FDA-approved allogeneic, cell-based therapies for Aging-related Frailty, Alzheimers Disease, or other aging-related conditions, or for HLHS or other cardiac-related indications; ethical and other concerns surrounding the use of stem cell therapy or human tissue; our exposure to product liability claims arising from the use of our product candidates or future products in individuals, for which we may not be able to obtain adequate product liability insurance; the adequacy of our trade secret and patent position to protect our product candidates and their uses: others could compete against us more directly, which could harm our business and have a material adverse effect on our business, financial condition, and results of operations; if certain license agreements are terminated, our ability to continue clinical trials and commercially market products could be adversely affected; the inability to protect the confidentiality of our proprietary information, trade secrets, and know-how; third-party claims of intellectual property infringement may prevent or delay our product development efforts; intellectual property rights do not necessarily address all potential threats to our competitive advantage; the inability to successfully develop and commercialize our product candidates and obtain the necessary regulatory approvals; we cannot market and sell our product candidates in the U.S. or in other countries if we fail to obtain the necessary regulatory approvals; final marketing approval of our product candidates by the FDA or other regulatory authorities for commercial use may be delayed, limited, or denied, any of which could adversely affect our ability to generate operating revenues; we may not be able to secure and maintain research institutions to conduct our clinical trials; ongoing healthcare legislative and regulatory reform measures may have a material adverse effect on our business and results of operations; if we receive regulatory approval of Lomecel-B or any of our other product candidates, we will be subject to ongoing regulatory requirements and continued regulatory review, which may result in significant additional expense; being subject to penalties if we fail to comply with regulatory requirements or experience unanticipated problems with our therapeutic candidates; reliance on third parties to conduct certain aspects of our preclinical studies and clinical trials; interim, topline and preliminary data from our clinical trials that we announce or publish from time to time may change as more data become available and are subject to audit and verification procedures that could result in material changes in the final data; the volatility of price of our Class A common stock; we could lose our listing on the Nasdaq Capital Market; provisions in our certificate of incorporation and bylaws and Delaware law might discourage, delay or prevent a change in control of our company or changes in our management and, therefore, depress the market price of our Class A common stock; we have never commercialized a product candidate before and may lack the necessary expertise, personnel and resources to successfully commercialize any products on our own or together with suitable collaborators; and in order to successfully implement our plans and strategies, we will need to grow our organization, and we may experience difficulties in managing this growth. Further information relating to factors that may impact the Companys results and forward-looking statements are disclosed in the Companys filings with the Securities and Exchange Commission, including Longeverons Annual Report on Form 10-K for the year ended December 31, 2023, filed with the Securities and Exchange Commission on February 27, 2024, as amended by the Annual Report on Form 10-K/A filed March 11, 2024, its Quarterly Reports on Form 10-Q, and its Current Reports on Form 8-K. The forward-looking statements contained in this press release are made as of the date of this press release, and the Company disclaims any intention or obligation, other than imposed by law, to update or revise any forward-looking statements, whether as a result of new information, future events, or otherwise.

Investor Contact: Derek Cole Investor Relations Advisory Solutions derek.cole@iradvisory.com

Photos accompanying this announcement are available at

https://www.globenewswire.com/NewsRoom/AttachmentNg/cfb4911d-2b0b-4c52-b2fa-b6982947155d

https://www.globenewswire.com/NewsRoom/AttachmentNg/e9e96d9c-d6c3-4aec-ba85-36631b520211

https://www.globenewswire.com/NewsRoom/AttachmentNg/f106bde7-972a-4687-a91e-03e924f409fc

Lomecel-B is a proprietary, scalable, allogeneic cellular investigational therapy developed by Longeveron for treating conditions like mild Alzheimers Disease.

The RMAT designation expedites the development and review process of Lomecel-B, providing benefits like FDA guidance and potential priority review, indicating its potential to address unmet medical needs in Alzheimers Disease.

The CLEAR MIND Phase 2a trial showed that Lomecel-B slowed/prevented the worsening of Alzheimers Disease compared to placebo, achieving primary safety and secondary efficacy endpoints.

Lomecel-B demonstrated in clinical trials the ability to slow or prevent the progression of Alzheimers Disease, suggesting it could be a viable therapeutic option for patients.

The full results of the CLEAR MIND Phase 2a trial will be presented at the Alzheimer's Association International Conference (AAIC) on July 28, 2024.

Read more here:
Longeveron Announces U.S. FDA Grants Lomecel-B Regenerative Medicine Advanced Therapy (RMAT) Designation for the Treatment of Mild Alzheimers Disease...

Posted in Regenerative Medicine | Comments Off on Longeveron Announces U.S. FDA Grants Lomecel-B Regenerative Medicine Advanced Therapy (RMAT) Designation for the Treatment of Mild Alzheimers Disease…

DiscGenics Announces the International Journal of Spine Surgery Has Published Results from an FDA-Approved Study of an Allogeneic Disc Progenitor Cell…

Posted: July 11, 2024 at 2:42 am

SALT LAKE CITY, July 9, 2024 /PRNewswire/ --DiscGenics, Inc., a privately held, late-stage clinical, biopharmaceutical company developing allogeneic, cell-based, regenerative therapies for musculoskeletal degeneration, today announced publication of results in the International Journal of Spine Surgery from the combined Phase I/Phase II, first-in-human clinical study of an allogeneic disc progenitor cell therapy (IDCT or rebonuputemcel) for painful lumbar degenerative disc disease (DDD).

The study met the primary safety and efficacy endpoints, showing that a single intradiscal injection of high-dose IDCT (9,000,000 cells/mL) safely increases disc volume and produces statistically significant, clinically meaningful improvements in back pain, disability, and quality of life out to 2 years post-injection in patients with lumbar disc degeneration.

"The results from this study demonstrate IDCT's potential to safely and effectively reduce pain associated with DDD while also producing a regenerative effect within the degenerating disc. MRI image analysis of disc volume indicated the potential to halt and possibly reverse the progression of the disease," said Matthew F. Gornet, M.D., lead author, Board Certified Spine Surgeon at The Orthopedic Center of St. Louis, and top enroller in the IDCT study. "I have been a practicing spine surgeon for more than 30 years and been involved in over 35 FDA clinical trials, and the patient outcomes from this study are very promising."

In the FDA-allowed, prospective, randomized, double-blinded, vehicle- and placebo-controlled,multicenter study, 60 patients with symptomatic, single-level lumbar DDD were randomized to receive single intradiscal injections of either low-dose cells (n= 20), high-dose cells (n= 20), vehicle alone (n= 10), or placebo (n= 10). The primary endpoint was mean visual analog scale (VAS) pain improvement >30% at 52 weeks. Disability and quality of life were evaluated via Oswestry Disability Index (ODI) and EQ-5D, respectively. Disc volume was radiologically assessed. Adverse events (AEs), regardless of whether they were related to treatment, were reported. Patients were assessed at baseline and at 4, 12, 26, 52, 78, and 104 weeks post treatment.

At Week 52, the primary study period, the high-dose group had a mean VAS percentage decrease from baseline (62.8%, P= 0.0005), achieving the endpoint of back pain improvement >30%; the mean change was also significantly greater than the minimal clinically important difference (MCID) of a 20-point decrease (42.8,P= 0.001). This clinical improvement was maintained at Week 104. In addition, the high-dose group had clinically meaningful, statistically significant improvements in ODI and EQ-5D by 12 weeks. Clinical improvement was sustained at 26 weeks, 52 weeks, 78 weeks, and 104 weeks following a single intradiscal injection. Only the high-dose group had a significant change in disc volume, with mean increases of 249.0 mm3(P= 0.028) at 52 weeks and 402.1 mm3(P= 0.028) at 104 weeks. Overall, a minority of patients (18.3%) reported AEs that were severe, with the highest percentage being reported in the placebo group. During the course of the trial, 6.7% of patients experienced serious AEs, all occurring in the vehicle (n= 1) or placebo (n= 3) groups, none were treatment related.

"Since the inception ofDiscGenics, we have seen consistent evidence of safety and the regenerative potential of the unique disc cell population in IDCT to address disc degeneration," said Kevin T. Foley, MD, Chief Medical Officer of DiscGenics and Chairman of Semmes-Murphey Neurologic & Spine Institute. "Our early basic science studies, our demonstrated ability to safely use human cells in 14 different pre-clinical animal studies conducted in the U.S.and Japan, and recently, data from our first-in-human safety and patient-reported outcomes published in this IJSS manuscript, all support the notion that this cell has the potential to safely regenerate the intervertebral disc from the inside-out."

DDD is a chronic and progressive condition where the intervertebral disc breaks down and causes pain and disability. It accounts for nearly 40% of chronic low back pain cases in the U.S., a serious medical condition that affects 12-30% of U.S. adults at a given time and is estimated to cost the U.S. healthcare system over $100 billion each year, creating a considerable burden on the economy and individual patients.

"The significant and durable results from this study demonstrate the incredible potential ofIDCT to change the paradigm of care for patients with DDD, a condition with limited treatment options," said Flagg Flanagan, Chief Executive Officer and Chairman of the Board for DiscGenics." We are excited by the momentum the publication of these study results offers, as we expect to initiate Phase III clinical study of this novel therapy in the U.S. imminently."

AboutIDCT

IDCT (injectable disc cell therapy, orrebonuputemcel) is a standalone, single-injection biologic treatment designed to halt progression of symptomatic lumbar disc degeneration and regenerate the disc from the inside out. The active ingredient (Drug Substance) in IDCT is a live, manufactured progenitor cell population derived from donated adult human intervertebral disc tissue. These cells are enriched and expanded into Discogenic Cells through a multistep manufacturing process in a highly controlled environment under current good manufacturing practices (cGMP) that results in significant proliferation and phenotypic changes to the cells. At the completion of the manufacturing process, the Discogenic Cells are subjected to extensive testing prior to use, including identity, purity, potency, and safety evaluations. The Discogenic Cells are then mixed with a viscous Sodium Hyaluronate Solution and excipients to generate IDCT, the Final Drug Product. IDCT is cryopreserved and maintained as individual "off-the-shelf" doses for administration viapercutaneous injection into the intervertebral disc in an outpatient setting. IDCT has been granted regenerative medicine advanced therapy (RMAT)and Fast Trackdesignations by the U.S. Food and Drug Administration.

Disclaimer:IDCT is an investigational product that is under development by DiscGenics and has not been approved by the FDA or any other regulatory agency for human use.

AboutDiscGenics

DiscGenics is a privately held, late-stage clinical biopharmaceutical company developingallogeneic, cell-based regenerative therapies for musculoskeletal degeneration. It's lead product candidate, IDCT (injectable disc cell therapy, or rebonuputemcel), is a standalone, single-injection biologic treatment designed to halt progression of lumbar disc degeneration and regenerate the disc from the inside out. DiscGenics is also developing a follow-on allogeneic cell platform to enable new musculoskeletal indications. To further development of these unique therapies, and to maintain control over compliance, cost, and production timelines, DiscGenics has built and validated an in-house scalable, allogeneic cell manufacturing process and cGMP facility at its headquarters in Salt Lake City, Utah. For more information, visitdiscgenics.com.

SOURCE DISCGENICS, INC.

Read the rest here:
DiscGenics Announces the International Journal of Spine Surgery Has Published Results from an FDA-Approved Study of an Allogeneic Disc Progenitor Cell...

Posted in Regenerative Medicine | Comments Off on DiscGenics Announces the International Journal of Spine Surgery Has Published Results from an FDA-Approved Study of an Allogeneic Disc Progenitor Cell…

Neha Motwani Elected to Longeveron Board of Directors – GlobeNewswire

Posted: July 11, 2024 at 2:42 am

MIAMI, July 09, 2024 (GLOBE NEWSWIRE) -- Longeveron Inc. (NASDAQ: LGVN), a clinical stage regenerative medicine biotechnology company developing cellular therapies for life-threatening and chronic aging-related conditions, including hypoplastic left heart syndrome (HLHS) and Alzheimers disease, today announced that Neha Motwani, a senior healthcare investment banker, has been elected to the Longeveron Board of Directors at the Companys recent Annual Meeting of Stockholders.

I am delighted to welcome Neha, with her tremendous healthcare industry experience, to the Board of Directors, said Joshua Hare, Co-founder, Chief Science Officer and Chairman of the Board at Longeveron. Her extensive knowledge of biopharma company operations, financing and capital markets will bring significant value to Longeveron as we continue to advance Lomecel-B, our proprietary, scalable, allogeneic cellular therapy. With five positive clinical trials across three indications, we believe Lomecel-Bhas the potential to be an important therapy for some of the most difficult diseases and conditions associated with aging.

Neha Motwani has over 25 years of healthcare investment banking experience, most recently serving as Managing Director, Healthcare Investment Banking at William Blair. She previously held investment banking roles of increasing responsibility with Truist Securities, Oppenheimer and Company, Stifel Financial and Cowen and Company, where, collectively, she completed transactions raising approximately $7.0 billion. Ms. Motwani earned her B.A. in political science from Columbia University.

I am delighted to join the Board of Directors of Longeveron at this exciting phase of the Companys clinical development, said Ms. Motwani. I look forward to working with this talented team to continue their mission to advance their novel cellular therapy, Lomecel-Bfor the potential treatment of HLHS, a rare and devastating congenital heart condition, and diseases of the aging.

About Longeveron Inc.

Longeveron is a clinical stage biotechnology company developing regenerative medicines to address unmet medical needs. The Companys lead investigational product is Lomecel-B, an allogeneic medicinal signaling cell (MSC) therapy product isolated from the bone marrow of young, healthy adult donors. Lomecel-B has multiple potential mechanisms of action encompassing pro-vascular, pro-regenerative, anti-inflammatory, and tissue repair and healing effects with broad potential applications across a spectrum of disease areas. Longeveron is currently pursuing three pipeline indications: hypoplastic left heart syndrome (HLHS), Alzheimers disease, and Aging-related Frailty. The Lomecel-BHLHS program has received three distinct and important FDA designations: Orphan Drug designation, Fast Track designation, and Rare Pediatric Disease designation. For more information, visit http://www.longeveron.com or follow Longeveron on LinkedIn, X, and Instagram.

Investor Contact: Derek Cole Investor Relations Advisory Solutions derek.cole@iradvisory.com

A photo accompanying this announcement is available at https://www.globenewswire.com/NewsRoom/AttachmentNg/00f0eb5c-76f5-49b1-9994-ee363513547b

Read this article:
Neha Motwani Elected to Longeveron Board of Directors - GlobeNewswire

Posted in Regenerative Medicine | Comments Off on Neha Motwani Elected to Longeveron Board of Directors – GlobeNewswire

Gene Therapy Pioneer Roger Hajjar, MD, Elected to Longeveron Board of Directors – GlobeNewswire

Posted: July 11, 2024 at 2:42 am

MIAMI, July 08, 2024 (GLOBE NEWSWIRE) -- Longeveron Inc. (NASDAQ: LGVN), a clinical stage regenerative medicine biotechnology company developing cellular therapies for life-threatening and chronic aging-related conditions, today announced that Roger Hajjar, MD, Director of the Gene and Cell Therapy Institute at Mass General Brigham, has been elected to the Longeveron Board of Directors. Dr. Hajjar is an internationally recognized scientist whose cardiac gene therapy discoveries have spurred clinical trials for heart failure, whose methodologies for cardiac-directed gene transfer are currently utilized by investigators around the world, and who was appointed as the inaugural director of the Gene and Cell Therapy Institute. Dr. Hajjar was elected at the Companys recent Annual Meeting of Stockholders.

I am delighted to welcome Roger, and his tremendous experience as a scientist, academic, and operational executive, to the Board of Directors, said Joshua Hare, Co-founder, Chief Science Officer and Chairman of the Board at Longeveron. His thought leadership in gene and cell therapy will bring significant value to Longeveron as we continue to advance Lomecel-B, our proprietary, scalable, allogeneic cellular therapy being evaluated in multiple indications. I look forward to collaborating with him to advance these important therapeutic development programs.

I am excited to join the Longeveron Board at this important time, both for the Company and in the history of gene therapy, said Dr. Hajjar. Gene and cell therapy is revolutionizing patient care with the potential to cure diseases or halt their progression. The Lomecel-Bdata generated to date in HLHS and Alzheimers disease support broad potential for this novel cellular therapy as a regenerative medical therapy across multiple indications and position the Company to potentially profoundly impact affected patients lives for the better.

Dr. Hajjars addition to the Board is part of the planned Board refreshment process at Longeveron, with a focus on bringing in new, relevant, experienced leaders over time to add to the knowledge base and experience provided by current and departing Board members.

Dr. Hajjar is an internationally renowned scientific leader in the field of cardiac gene therapy. His laboratory had validated a number of cardiac targets that led to the initiation and completion under his guidance of First-in-Human gene therapy trials in patients with heart failure. He has initiated multiple clinical trials in gene therapy for a variety of cardiovascular diseases.

He has authored over 500 publications and has received numerous awards for his achievements in the field of cardiac gene therapy. Dr. Hajjar received his Bachelor of Science in Biomedical Engineering from Johns Hopkins University and his Doctor of Medicine from Harvard Medical School and the Harvard-MIT Division of Health Sciences & Technology. He completed his training in internal medicine, cardiology, heart failure/cardiac transplantation, and research fellowships at Massachusetts General Hospital in Boston where he then directed the Cardiology Laboratory of Integrative Physiology and Imaging. From 2007 to 2018, Dr. Hajjar was the Director of the Cardiovascular Research Center, and the Arthur & Janet C. Ross Professor of Medicine at Mount Sinai School of Medicine in New York. From 2019 to 2022, Dr. Hajjar was involved in the creation of a number of gene therapy companies at Flagship Pioneering, Cambridge, MA and he was the head of R&D at Ring Therapeutics.

About Longeveron Inc.

Longeveron is a clinical stage biotechnology company developing regenerative medicines to address unmet medical needs. The Companys lead investigational product is Lomecel-B, an allogeneic medicinal signaling cell (MSC) therapy product isolated from the bone marrow of young, healthy adult donors. Lomecel-B has multiple potential mechanisms of action encompassing pro-vascular, pro-regenerative, anti-inflammatory, and tissue repair and healing effects with broad potential applications across a spectrum of disease areas. Longeveron is currently pursuing three pipeline indications: hypoplastic left heart syndrome (HLHS), Alzheimers disease, and Aging-related Frailty. The Lomecel-BHLHS program has received three distinct and important FDA designations: Orphan Drug designation, Fast Track designation, and Rare Pediatric Disease designation. For more information, visit http://www.longeveron.com or follow Longeveron on LinkedIn, X, and Instagram.

Investor Contact: Derek Cole Investor Relations Advisory Solutions derek.cole@iradvisory.com

A photo accompanying this announcement is available at https://www.globenewswire.com/NewsRoom/AttachmentNg/0ff5c44e-e63d-4909-b8b8-6d71186087ec

Excerpt from:
Gene Therapy Pioneer Roger Hajjar, MD, Elected to Longeveron Board of Directors - GlobeNewswire

Posted in Regenerative Medicine | Comments Off on Gene Therapy Pioneer Roger Hajjar, MD, Elected to Longeveron Board of Directors – GlobeNewswire

Treating heart attacks with stem cells: The ‘biggest breakthrough’ yet? – The Week

Posted: July 11, 2024 at 2:42 am

Here's one way to mend a broken heart: Researchers from the University of Louisville, Kentucky, used cardiac stem cells to repair the damaged heart muscles of patients who had experienced severe heart attacks. Some call the findings, which were published in the journal Lancet, the field's "biggest breakthrough in a generation." Here's what you should know:

Wait, this treatment helps after a heart attack?

It does. Usually, the recovery phase after a heart attack is dangerous because of the possibility of heart failure, says Britain's Telegraph. Heart failure which is often linked with heart attack victims, but can strike other people, too "occurs when a damaged heart is weakened and unable to pump enough blood around the body." This treatment helps reduce that risk.

Escape your echo chamber. Get the facts behind the news, plus analysis from multiple perspectives.

SUBSCRIBE & SAVE

From our morning news briefing to a weekly Good News Newsletter, get the best of The Week delivered directly to your inbox.

From our morning news briefing to a weekly Good News Newsletter, get the best of The Week delivered directly to your inbox.

How does the treatment work?

It uses cardiac stem cells taken directly from the heart to repair the organ. "Cardiac stem cells can renew themselves and can form one of three types of cell that make up heart tissue," says Jessica Hamzelou at New Scientist. Researchers hypothesized that injecting stem cells directly into damaged areas might help improve the heart's recovery after an attack. In this new study, researchers extracted stem cells from the hearts of 16 patients during bypass surgery, then "multiplied them in culture and injected them back into each original patient's heart" four months later.

And this helped them recover?

It did. When controlled against patients who received standard care, the 16 patients demonstrated "striking" improvements. Their "pumping efficiency" was noticeably better, says the Telegraph, and surgical scars even shrank. After a year, eight of the patients saw a 12 percent improvement in pumping efficiency triple what researchers initially expected.

A free daily email with the biggest news stories of the day and the best features from TheWeek.com

Why is this such a big deal?

"It's long been an article of faith that human heart muscle, once damaged, cannot regenerate," says Eryn Brown at the Los Angeles Times. This study proves that not only can the heart be repaired, but that with a little assistance, it can repair itself using its own cells. That's very good news, as an estimated 6 million people in the U.S. experience heart failure every year.

Sources: LA Times, New Scientist, Telegraph

To continue reading this article...

Create a free account

Continue reading this article and get limited website access each month.

Already have an account? Sign in

Subscribe to The Week

Get unlimited website access, exclusive newsletters plus much more.

Cancel or pause at any time.

Already a subscriber to The Week?

Unlimited website access is included with Digital and Print + Digital subscriptions. Create an account with the same email registered to your subscription to unlock access.

Follow this link:
Treating heart attacks with stem cells: The 'biggest breakthrough' yet? - The Week

Posted in Kentucky Stem Cells | Comments Off on Treating heart attacks with stem cells: The ‘biggest breakthrough’ yet? – The Week

How the Hidden Mechanics of Genetic Medicines Can Disadvantage Non-White Individuals – Leonard Davis Institute

Posted: July 11, 2024 at 2:42 am

Carmen Guerra, LDI Senior Fellow and Vice Chair of Diversity and Inclusion in the Perelman School of Medicines Department of Medicine warned of health care disparities driven by the limitations of some genetic-based immunological drugs.

As a panelist at the White House Minority Health Forum, LDI Senior Fellow Carmen Guerra, MD, MSCE, warned of the equity implications of genetic-based immunological drugs that are dependent on a genetic element that is prominent in white populations but not in other populations.

Guerra is Vice Chair of Diversity and Inclusion in the Perelman School of Medicines Department of Medicine and Associate Director of Diversity and Inclusion at the University of Pennsylvanias Abramson Cancer Center. As a nationally recognized expert in the field of health and health care disparities, she was invited to be a panelist on the White House Office of Science and Technology Policys inaugural White House Minority Health Forum.

The event was organized to recognize National Minority Health Month and to highlight progress, discuss challenges, and identify actions that the federal government and private sector can take to improve health outcomes and reduce health inequities for racial and ethnic minority communities across the country.

Setting the tone of the forum sessions, Director of the Office of Science and Technology Policy Arati Prabhakar said, Our health outcomes in America are simply unacceptable for the richest country in the world and that is especially true in our ethnic minority communities and our racial minority communities.

Guerra was a panelist in the Research Innovation for Health Equity session moderated by Cameron Webb, MD, JD, Director of Health Policy and Equity at the University of Virginia School of Medicine and former senior advisor to the White House Covid-19 Response Team.

He asked Guerra, What are we missing in our current paradigm in terms of how to effectively study how to create opportunities for everybody to achieve their best health?

One area is genomic data and how it lacks representation for all groups, said Guerra. Another example is the immunotherapy field that is exploding with new groups of therapies for many diseases, including cancer. Some really interesting recent research shows that many of the targeted immunological therapies being developed rely on a specific HLA allele that is actually very prominent in most white populations in the Americas and Europe but doesnt represent the rest of the population of the world.

She said that racial health disparity is going to get worse because about 80% of the therapies now being studied are specific for that allele in Anglo populations.

An HLA allele is a variant form of a gene within the human leukocyte antigen (HLA) system involved in regulating the immune system. A recent commentary in the Journal for ImmunoTherapy of Cancer titled Ensuring Equity in the Era of HLA-Restricted Cancer Therapeutics, noted that a new melanoma-targeting drug approved in 2022 by the U.S. Food and Drug Administration is dependent on an HLA allele whose expression varies considerably among ethnic groups. It is most frequently expressed in Europeans, and less commonly in African Americans and people of Asian or Pacific Island ancestry. We advocate for proactive consideration of the populations eligible for each HLA-restricted therapeutic in development to ensure this emerging therapeutic class does not compound long-standing health disparities.

Blog Post

Health Care Access & Coverage

Using Nudges Like Text Messages Can Help Make Public Health Gains

News

Health Equity

A Widespread Presence at the Countrys Largest Gathering of Health Services Researchers

News

Health Equity

Penn LDI Health Services Researchers Spotlight and Network Around Their Latest Work

In Their Own Words

Health Care Access & Coverage

Evidence Points to Health Improvements Linked to Tax Credit Payments

News

Health Equity

Baltimore Event Immerses Them in the National Health Services Research Community

Go here to see the original:
How the Hidden Mechanics of Genetic Medicines Can Disadvantage Non-White Individuals - Leonard Davis Institute

Posted in Genetic medicine | Comments Off on How the Hidden Mechanics of Genetic Medicines Can Disadvantage Non-White Individuals – Leonard Davis Institute

Ellen DeGeneres Says Shes Done With Fame After Netflix Special Drops – The Daily Beast

Posted: July 11, 2024 at 2:42 am

Bryan Johnson, the billionaire tech CEO whose sole goal in life seems to be to live forever, underwent an experimental longevity treatment in the Bahamas last month that involved pumping himself up with 300 million stem cellsand documented it all for his YouTube channel. Johnson, 46, flew out to the Albany, a high-end Bahamas golf resort co-owned by the likes of Tiger Woods and Justin Timberlake, for the procedure. The stem cells, sourced from bone marrow by a company called Cell Colars Clinical, target a patients joints in the hopes of rejuvenating them, Johnson explained in a June 25 vlog. Those healthy young Swedish cells should multiply in my body, future-proofing all of my major joints and taking me one step closer to age 18, he said. The entrepreneur, who all but abandoned Silicon Valley for biohacking and a strict $2-million-a-year regimen he calls Project Blueprint, claimed earlier this year to have reversed his epigenetic age by a little over five years.

Read more:
Ellen DeGeneres Says Shes Done With Fame After Netflix Special Drops - The Daily Beast

Posted in Stem Cells | Comments Off on Ellen DeGeneres Says Shes Done With Fame After Netflix Special Drops – The Daily Beast

LRIG1 controls proliferation of adult neural stem cells by facilitating TGF and BMP signalling pathways – Nature.com

Posted: July 11, 2024 at 2:42 am

Mice

Lrig1 constitutive knockout mice were generated by crossing Lrig1CreERT2 (Jackson Strain No. 018418, Lrig1tm1.1(cre/ERT2)Rjc/J) heterozygous mice on the 129-Elite strain background (Charles River Canada, strain code 476) together to produce homozygous knockout mice as described in ref. 14. Genotypes were confirmed post-weaning from ear notch genomic DNA (isolated with DirectPCR Lysis Reagent (Ear, Cat # 401-E)) using PCR primers and Jackson Protocol 26202 exactly as described. For all experiments female and male mice were randomly assigned to experimental groups and analyzed at ages of 8-weeks, 15-weeks and 24-weeks. Specific timepoints are noted in the figure legends and captions. Mice used in this work all followed a 12h light/dark cycle and had ad libitum access to water and rodent chow. Mice did not demonstrate any visible signs of being immunocompromised or display any obvious behavioural phenotypes. All animal work conducted followed policies formed by the Canadian Council of Animal Care and approved by the Local Animal Care Committee at the University of Toronto and The Hospital for Sick Children.

Frozen tissue sections were dried for 1h at room temperature or 30min at 37C, then rehydrated in phosphate buffered saline (PBS, 137mM NaCl, 2.7mM KCl, 10mM Na2HPO4, 1.8mM KH2PO4) for 10min. Subsequently, sections were blocked and permeabilized in a solution of 5% BSA and 0.3% Triton X-100 in PBS. Following blocking, sections were incubated in primary antibody diluted in 1:1 PBS to blocking solution overnight, at 4C in a humidified chamber. The following day sections were washed in PBS three times for 5min, then incubated in diluted secondary antibody for 2h at room temperature. Afterwards, sections were washed three times in PBS for 5min per wash. Finally, sections were counterstained for 5min at room temperature in 0.5g/mL Hoechst 33258 (Sigma-Aldrich), washed with PBS and then mounted with PermaFluor mountant (Thermo Scientific).

5-ethynyl-2-deoxyuridine (EdU, Toronto Research Chemicals Cat #: E932175) was dissolved in PBS and injected intraperitoneally three times two hours apart at a dose of 50mg/kg to 15-week-old Lrig1 WT and KO mice. After 3 weeks brains were collected and prepared for IHC as described below. EdU+ cells were detected using the Click-it EdU Alexa 488 kit (Invitrogen, Cat #: C10337) following the manufacturers instructions and then stained with Hoechst 33258. EdU+ cells were counted from five sections per brain.

Tissue sections from 15-week-old Lrig1 WT and KO mice were prepared as described below, blocked and incubated overnight with 1:100 anti-LRIG1 and anti-BMPR-1B (1:100 see Supplementary TableS3) antibodies as described above for IHC. The next day sections were washed three times for 10min with Wash Buffer A (0.01M Tris, 0.15M NaCl and 0.05% Tween-20) and then incubated with the Duolink In Situ PLA Probe Anti-Goat

PLUS (Sigma-Aldrich) and Duolink In Situ PLA Probe Anti-Mouse MINUS Affinity purified Donkey anti-mouse IgG (H+L) (Sigma-Alrich, See Supplementary TableS4) for 1hour at 37 C in a humidified chamber. Following washing of the PLA probes, Ligation and Amplification was carried out as described by the manufacturer using the Duolink In Situ Detection Reagents kit (Sigma-Aldrich) and nuclei stained for Hoechst 33258.

The periventricular area containing the V-SVZ from N=2 female and N=2 male Lrig1 KO and N=3 female and N=1 male WT 8-week old brains was dissected as described in ref.18 from both hemispheres of each brain and stored at 80C until needed. One hemisphere sample was used to isolate RNA as described below. The other hemisphere sample was lysed and western blotted for LRIG1 and pEGFR as described previously14.

Lrig1 KO and WT mice were anesthetized with 23% inhaled isoflurane and then perfused transcardially with PBS followed by 4% paraformaldehyde (PFA). Brains were then dissected, post-fixed overnight in 4% PFA and then cryoprotected in 30% sucrose in PBS for 24h. Tissues were then embedded with O.C.T (Fisher Healthcare Tissue-Plus O.C.T. Compound, Cat # 23-730-571) and sections were cut 18m thick in the coronal plane using a Thermo Fisher Scientific HM525 NX cryostat at 20C. Sections were collected on glass slides (Fisherbrand Superfrost Plus Microscope Slides, Cat #1255015) coated with gelatin and stored frozen until use.

All primary and secondary antibodies used for western blot are listed in Supplementary TablesS1 and S2, while those used for IHC/PLA are listed in Supplementary TablesS3 and S4.

For all experiments, plasmid constructs were used following endotoxin-free maxipreps using a Qiagen EndoFree Plasmid Maxi Kit or a ZymoPURE II Plasmid Maxiprep Kit. Plasmid DNA concentration was determined using a NanoDrop 2000 (ThermoFisher). All plasmids used in this study are listed in Supplementary TableS5.

Images were collected using a Zeiss Spinning Disk confocal microscope system or a Zeiss AxioImager M2 microscope system with a Calibri LED light source. Images were acquired using Z-stacks (with the apotome engaged in the case of the AxioImager) and staked tiles imaging set-up using Zen Blue software. All images were acquired with Z-stack sizes ranging between 15-25 slices depending on the dataset analyzed. Images shown were produced using the orthogonal projection feature implemented in Zen Blue.

For all cell count analysis on acquired images from V-SVZ, counting was done using ImageJ. For cell counts in the V-SVZ, only visibly immunostained (positive) cells along the dorsal and ventral portions of the lateral wall (LW), closest to the ventricle (periventricular area) were counted. To differentiate between the dorsal and ventral areas of the LW, these regions were measured using the line drawing and measurement tools in ImageJ along the length of the entire LW. To assess proliferation, neuronal progeny and pEGFR-positive cells in the V-SVZ, the top 1/3rd of the length of the LW was considered the dorsal portion. The bottom 2/3 measurement of the LW was considered the ventral portion. To obtain a cell count value for each individual brain, positive cells counted from three anatomically matched 18 m thin coronal sections containing the V-SVZ LW region were totaled and averaged. For the proportion of proliferating (Ki67+) cells, a percent cell count was used to represent the data by dividing the number of SOX2+GFAP+Ki67+/SOX2+GFAP+ cells. For the number of DCX-positive and pEGFR-positive cells the total number of cells counted per ventricle area was used to represent these data. For cell counts in the OB, positive cells in the GCL, GL and MCL were counted using three anatomically 18 m thick coronal sections of the whole OB from each mouse brain. To create a cell count value for each individual brain, counts from all three sections were totaled and averaged. For the number of CalB or CalR-positive interneurons, the total number of cells in each layer were counted and used to represent these data. pSmad2 and pSmad1/5/9 positive cells only in the V-SVZ were counted on the entire ventral portion of the LW of the LV. Three coronal sections from each brain which were anatomically matched in order of contain the LW area of the V-SVZ and were used for cells count analysis, where the total positive cells from each coronal section were then averaged.

Following RNA isolation using a RNeasy Plus Mini Kit (Qiagen, Cat#:74134) from tissue isolated as described above, polyA selected mRNA next generation sequencing libraries were prepared using the NEBNext Ultra II DNA Library Prep Kit and sequenced on one lane of an Illumina Novaseq SP flow cell achieving ~4050 million reads per sample. Following sequencing, FASTQ files were generated with bcl2fastq2 v2.20. Library preparation, sequencing and FASTQ file production were done by The Centre for Applied Genomics (TCAG) at the Hospital for Sick Children. FASTQ files were then used as input for Salmon19 for alignment to the mouse genome (Gencode m29) and for read quantification. Deseq2 as implemented in R and was used for normalization and differential gene expression analysis20.

Immunoprecipitation (IP) assays were done similar to those described in ref. 21. Neuro-2a cells (N2a) (ATCC Cat No: CCL-131) were used for all IP experiments which were cultured in Dulbeccos modified Eagles medium (DMEM) containing high levels of glucose supplemented with 1% penicillin-streptomycin and 10% fetal bovine serum (FBS). Transfections of N2a cells were carried out using Poly-Jet reagent (SignaGen Laboratories; Cat# SL100688) 24h after plating 400,000 cells per well of 6 well plate. Flag-tagged pCMV expression vectors containing either the TGFR1, TGFRII and BMPRI were used to co-transfect N2a cells with Lrig1 pCMV-overexpression plasmid using PolyJet In Vitro DNA Transfection Reagent (SignaGen Laboratories; Cat# SL100688). A mock condition with no plasmids was used as a negative control and BMPRI was used as a positive control21. N2a cells were lysed after 48h in TNTE buffer containing 0.5% Triton-X-100 (150mM NaCl, 50mM Tris pH 7.4, and 1mM EDTA)21. Anti-Flag Magnetic beads (Selleckchem Cat#: B26101) were used to carry out all immunoprecipitations. The beads and sample mixtures were incubated on a tube rotator at 4C for 2h. Supernatant was then discarded and the beads were washed three times with wash buffer (TNTE buffer containing 0.1% Triton-X100) on a magnetic separation rack. Following the final wash, magnetic beads were eluted using 1x SDS-PAGE loading buffer heated at 70C for 10min. 10% of the samples prior to immunoprecipitation were collected for a load condition in 5x SDS Sample Loading Buffer (10% SDS, 500mM DTT, 50% Glycerol, 250nM Tris-HCl, 0.5% bromophenol blue dye, pH 6.8). Western blotting was performed as described previously14 using anti-flag antibody (DYKDDDDK Tag Antibody Cell Signalling Technology Cat#: 2368) to detect the TGFR1, TGFRII and BMPRI receptors and LRIG1 antibody (R&D Systems, Cat#:3688) to detect LRIG1. GAPDH antibody was used as an internal reference to normalize protein expression levels.

LRIG1 ectodomain (ECD, Cat#: 3688-LR-050 from R&D Systems) concentrations of 2.5g/mL, 0.25g/mL, and 0.025g/mL were immobilized on a nitrocellulose membrane in duplicate. 2.5% Bovine serum albumin and 1xPBS were used as negative controls. Once dried, the blot was incubated at room temperature (RT) in blocking buffer (2% BSA in PBS-T (1xPBS containing 0.1% Tween)) for 30min. 1.2 ug of biotinylated human TGF1 (Avi- Tag, Biotin-Labeled, BPS Bioscence Cat #. 100843) was then added to blocking buffer on the blot and incubated for one hour at RT. A 1:5000 dilution of Streptavidin-HRP in blocking buffer was added for 30min followed by 35min and 315-minute washes with PBS-T. The blot was then developed with Bio-Rad Clarity Western enhanced chemiluminescence (ECL) Detection Reagent on a Syngene G:Box chemiluminescence imager.

The binding affinity of LRIG1 extracellular domain (ECD) to TGF1 was measured using a BLItz instrument. LRIG1 ECD in 1xPBS was used as the analyte and biotinylated-TGF1 was used as the bait. Streptavidin-coated sensors were hydrated in BLI rehydration buffer (PBS, 0.5mg/mL BSA, and 0.02% (v/v) Tween-20). The biotinylated bait protein was diluted in kinetics buffer (PBS, 0.5mg/mL BSA, 0.02% (v/v) Tween-20) to a final concentration of 12.4g/mL. The bait was immobilized on a streptavidin-coated biosensor tip for 30s. Next, the analyte concentrations were diluted in kinetic buffer in order to obtain concentration of LRIG1 ECD of 1M, 750nM, 500nM, 100nM. The binding association was measured over a period of 120s with subsequent dissociation measured after immersion of the biosensor tip into kinetic buffer for another 120s. The data were analyzed and sensorgrams were step corrected, reference corrected and globally fit to a 1:1 binding model. Dissociation constants (KD) were calculated using BLItz Pro Version 1.1.0.16 and the average of two independent determinations is reported here.

N2a cells were transfected with guide RNAs #1, #2, or LacZ control contained in the pU6-(Bbsl)_CBh_Cas9_T2A_mCherry plasmid (as described previously14 in using Lipofectamine STEM, according to manufacturers protocol in 6 well plates. Two days following transfection, cells were replated into 96 well plates at a concentration of 0.5 cells/200l media in each well. Cells were allowed to grow for 3 weeks and wells containing individual colonies that expressed red fluorescence from mCherry were selected and propagated further to produce clonal cell lines. Three control lines and four LRIG1 KO lines were selected following western blotting for LRIG1 to confirm loss of the LRIG1 protein and used for the experiments described here.

In the case of TGF1 and BMP4 treated non-transfected Ctrl N2a clonal cell lines and Lrig1 KO N2a clonal cell line experiments, cells were plated at 90,000 cells/well in DMEM media (described above) in 24-well plates. Two days later the media was changed to DMEM containing 0.1% FBS for serum starvation overnight. The following day, 10ng/ml TGF1 (Cedarlane; Cat# 781802) or BMP4 (Peprotech Cat#315-27) ligands suspended in DMEM containing 0.1% FBS. The later of which on its own was used as the control. Following 30min at 37C, the cells were collected using 50L of lysis buffer containing, tris-buffered Saline, pH 7.4, 0.5% b-octyl-D-glucopyranoside, 0.5% Triton X-100, 1mM NaF, 1mM -glycerophosphate, 1mM Na3VO4 and 1 cOmplete, mini, EDTA-free protease inhibitor tablet per 10ml. Protein assay was done with the samples using the DC Assay (Bio-Rad) and then equal amounts of protein were used for western blots to assess the pSmad2/3, total Smad2, pSMAD1/5/9 and total SMAD1 along with GAPDH (as a loading control) and LRIG1 (to confirm genotypes).

Since N2a cells express low levels of TGFRII, we used TGFRI and TGFRII constructs to express these receptors in Ctrl and Lrig1 KO clonal N2a cells followed by treatment with TGF1 to assess the response of this signalling pathway in the context of loss of LRIG1. To do so, 1g of DNA per well, using PolyJet of TGFRI and TGFRII were transfected one day after plating 90,000 cells per well of each of the Ctrl and Lrig1 KO clonal N2a cell lines. 24h after transfection, the cells were transitioned into DMEM containing 0.1% FBS media. The following the Lrig1 KO and WT clonal cell lines were treated with 10ng/ml per well of TGF1 for 30min at 37C, lysed, and protein concentrations determined as above. Western blotting for pSmad2/3, total Smad2 and GAPDH were then performed.

Neurospheres were grown essentially as described in ref. 18 using NeuroCult Mouse&Rat Basal Medium containing NeuroCult Proliferation Supplement (STEMCELL Technologies), FGF2 (Corning), EGF (STEMCELL Technologies), 0.2% heparin (Sigma-Aldrich, Cat #: H4784) and 1% penicillin-streptomycin from brains of 15 week old Lrig1 WT and KO mixed male and female mice (N=2 male and N=1 female KO mice, N=2 male and N=2 female WT mice). Following seven days of growth of secondary spheres, spheres from each brain were gently distributed into two wells of a 24-well plate and treated with BMP4 as described above without changing the culture media. Cells were then collected by centrifugation and western blots were performed as described above.

Data previously published by Mizrak et al.22, from lateral wall cells of 810 week old mice was obtained from NCBI GEO accession number: GSE109447. Specifically, the 13,055 cell data set was reduced to just the lateral wall cells and then was processed using Seurat (version 4.4.0) as implemented in R (version 4.2.1). Cells with less than 200 genes expressed in at least three cells were removed. The data were then normalized and scaled, principal components were computed and clustering was performed with 25 principal components and a resolution of 0.8. UMAP plots were created using the DimPlot and FeaturePlot functions as implemented in Seurat.

GraphPad Prism Version 9.3.1 software was used to perform all statistical analyses and create graphical representations. For statistical analyses in Figs.13, a two-way ANOVA and Tukeys Honestly Significant Difference (HSD) post-hoc test were used to compare data between the different experimental groups present in each paradigm (ex. KO vs. WT), and the area of the LW being analyzed (ex. dorsal vs ventral). In Fig.5, a two-tailed Students t-test in order to compare the differences between Lrig1 KO and WT groups. In Fig.7 and Supplementary Fig.S3, the results from the four conditions were averaged from two independent experiments and normalized to total SMAD2/3 for the TGFR transfections or total SMAD1 for the BMP experiments. Subsequently, a one-way ANOVA followed by a dk post-hoc test was used to compare groups. In all figures, values were reported as means. To create error bars, means were represented as the standard error of the mean (S.E.M). Experimental data was denoted as statistically significant at P values less than 0.05 (P<0.05). In all the figures, *p<0.05, **p<0.01, ***p<0.001 and ns = not significant.

a Immunostaining of the lateral wall of 15-week old Lrig1 KO and WT mice for GFAP, SOX2, Ki67 and merged with Hoechst to label nuclei. Sox2/GFAP-double positive cells allows identification of NSCs. Ki67/SOX2/GFAP-triple positive cells represent proliferating NSCs. Arrowhead indicate triple positive cells. Quantification of the number of SOX2/GFAP-double positive NSCs (b) and proportion of proliferating Ki67-positive NSCs (c) along the dorsal and ventral portions of the lateral wall in 15-week old Lrig1 KO and WT mice. d Immunostaining of the lateral wall of 24-week old Lrig1 KO and WT mice for GFAP, SOX2, Ki67 and Merged with Hoechst. Quantification of the number of SOX2/GFAP-double positive NSCs (e) and proportion of proliferating Ki67-positive NSCs (f) along the dorsal and ventral portions of the lateral wall in 24-week old Lrig1 KO and WT mice. Scale bars represents 25m. Error bars indicate S.E.M. For (ac, N=5 per group, and for df, N=3 per group. Source data for graphs are included in Supplementary Data2.

a Immunostaining of coronal sections through the lateral ventricle of 15-week old Lrig1 KO and WT mice for DCX to mark new-born neurons/neuroblasts and GFAP to label ventricular cells merged with Hoechst. b Quantification of the number of DCX-positive cells in the dorsal and ventral portions of the lateral wall. c Representative immunostaining of Olfactory Bulb (OB) sections with antibodies for Calretinin (CalR) and Calbindin (CalB) and merged with Hoechst at low magnification (left, indicating the Granule Cell Layer (GCL), the Mitral Cell Layer (MCL) and the Glomerular Layer (GL)) and at high magnification (right) from Lrig1 and KO mice. d Quantification of the number of CalB-positive cells in each of the granule cell (GCL), mitral cell (MCL) or glomerular layers (GL), (e) Quantification of the number of CalB-positive cells in the GCL, MCL and GL of the OB. f, g Quantification of the number of EdU-positive cells in the olfactory bulb (f) and representative staining of EdU-positive cells with Hoechst from an Lrig1 WT brain. Scale bars represent 50m for (a), 100m for (c) and 20m for (g). Error bars represent S.E.M. For (a, b, N=5 per group and for cf, N=3 per group. Source data for graphs are included in Supplementary Data2.

a Immunostaining of coronal sections along the lateral wall of Lrig1 KO and WT mice with antibodies for phosphorylated EGFR (pEGFR) and merged with Hoechst. b Quantification of the number of pEGFR-positive cells in the dorsal and ventral portions of the lateral wall of the V-SVZ. c Western blots using antibodies for LRIG1, pEGFR and GAPDH (loading control) from periventricular tissue dissected from one hemisphere per animal from 8-week old Lrig1 KO and WT mice See also Supplementary Fig.S2d. d Quantification of the western blot data from (c) for pEGFR relative to GAPDH. Scale bar represents 50um. Error bars represent S.E.M. For (a, b, N=5 per group and for c, N=4 per group. Source data for graphs are included in Supplementary Data2.

See original here:
LRIG1 controls proliferation of adult neural stem cells by facilitating TGF and BMP signalling pathways - Nature.com

Posted in Stem Cells | Comments Off on LRIG1 controls proliferation of adult neural stem cells by facilitating TGF and BMP signalling pathways – Nature.com

Cancer stem cells: advances in knowledge and implications for cancer therapy – Nature.com

Posted: July 11, 2024 at 2:42 am

Siegel, R. L., Giaquinto, A. N. & Jemal, A. Cancer statistics, 2024. CA Cancer J. Clin. 74, 1249 (2024).

Article PubMed Google Scholar

Bonnet, D. & Dick, J. E. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat. Med. 3, 730737 (1997).

Article CAS PubMed Google Scholar

Batlle, E. & Clevers, H. Cancer stem cells revisited. Nat. Med. 23, 11241134 (2017).

Article CAS PubMed Google Scholar

Plaks, V., Kong, N. & Werb, Z. The cancer stem cell niche: how essential is the niche in regulating stemness of tumor cells? Cell Stem Cell 16, 225238 (2015).

Article CAS PubMed PubMed Central Google Scholar

Yang, L. et al. Targeting cancer stem cell pathways for cancer therapy. Signal. Transduct Target Ther. 5, 8 (2020).

Article PubMed PubMed Central Google Scholar

Houghton, J., Morozov, A., Smirnova, I. & Wang, T. C. Stem cells and cancer. Semin. Cancer Biol. 17, 191203 (2007).

Article CAS PubMed Google Scholar

Sell, S. Stem cell origin of cancer and differentiation therapy. Crit. Rev. Oncol. Hematol. 51, 128 (2004).

Article PubMed Google Scholar

Shimkin, M. B. The written word and cancersome personal involvements, 1940-1977: autobiographical essay. Cancer Res. 38, 241252 (1978).

CAS PubMed Google Scholar

Cairns, J. Mutation selection and the natural history of cancer. Nature 255, 197200 (1975).

Article CAS PubMed Google Scholar

Fisher, J. C. & Hollomon, J. H. A hypothesis for the origin of cancer foci. Cancer 4, 916918 (1951).

Article CAS PubMed Google Scholar

Furth, J., Kahn, M. C. & Breedis, C. The transmission of Leukemia of Mice with a Single Cell1. Am. J. Cancer 31, 276282 (1937).

Google Scholar

Till, J. E. & Mc, C. E. A direct measurement of the radiation sensitivity of normal mouse bone marrow cells. Radiat Res. 14, 213222 (1961).

Article CAS PubMed Google Scholar

Siminovitch, L., McCulloch, E. A. & Till, J. E. The distribution of colony-forming cells among spleen colonies. J. Cell Comp. Physiol. 62, 327336 (1963).

Article CAS PubMed Google Scholar

Potten, C. S. & Loeffler, M. Stem cells: attributes, cycles, spirals, pitfalls and uncertainties. Lessons for and from the crypt. Development 110, 10011020 (1990).

Article CAS PubMed Google Scholar

Orkin, S. H. & Zon, L. I. Hematopoiesis: an evolving paradigm for stem cell biology. Cell 132, 631644 (2008).

Article CAS PubMed PubMed Central Google Scholar

Schofield, R. The relationship between the spleen colony-forming cell and the haemopoietic stem cell. Blood Cells 4, 725 (1978).

CAS PubMed Google Scholar

Zhang, J. et al. Identification of the haematopoietic stem cell niche and control of the niche size. Nature 425, 836841 (2003).

Article CAS PubMed Google Scholar

Jassim, A., Rahrmann, E. P., Simons, B. D. & Gilbertson, R. J. Cancers make their own luck: theories of cancer origins. Nat. Rev. Cancer 23, 710724 (2023).

Article CAS PubMed Google Scholar

Riva, L. et al. The mutational signature profile of known and suspected human carcinogens in mice. Nat. Genet. 52, 11891197 (2020).

Article CAS PubMed PubMed Central Google Scholar

Soto, A. M. & Sonnenschein, C. The tissue organization field theory of cancer: a testable replacement for the somatic mutation theory. Bioessays 33, 332340 (2011).

Article PubMed PubMed Central Google Scholar

Reya, T., Morrison, S. J., Clarke, M. F. & Weissman, I. L. Stem cells, cancer, and cancer stem cells. Nature 414, 105111 (2001).

Article CAS PubMed Google Scholar

Clarke, M. F. Clinical and therapeutic implications of cancer stem cells. N Engl. J. Med. 380, 22372245 (2019).

Article CAS PubMed Google Scholar

Al-Hajj, M. et al. Prospective identification of tumorigenic breast cancer cells. Proc. Natl Acad. Sci. USA 100, 39833988 (2003).

Article CAS PubMed PubMed Central Google Scholar

Singh, S. K. et al. Identification of human brain tumour initiating cells. Nature 432, 396401 (2004).

Article CAS PubMed Google Scholar

Matsui, W. et al. Characterization of clonogenic multiple myeloma cells. Blood 103, 23322336 (2004).

Article CAS PubMed Google Scholar

Collins, A. T. et al. Prospective identification of tumorigenic prostate cancer stem cells. Cancer Res. 65, 1094610951 (2005).

Article CAS PubMed Google Scholar

Fang, D. et al. A tumorigenic subpopulation with stem cell properties in melanomas. Cancer Res. 65, 93289337 (2005).

Article CAS PubMed Google Scholar

OBrien, C. A., Pollett, A., Gallinger, S. & Dick, J. E. A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature 445, 106110 (2007).

Article PubMed Google Scholar

Ricci-Vitiani, L. et al. Identification and expansion of human colon-cancer-initiating cells. Nature 445, 111115 (2007).

Article CAS PubMed Google Scholar

Li, C. et al. Identification of pancreatic cancer stem cells. Cancer Res. 67, 10301037 (2007).

Article CAS PubMed Google Scholar

Rahman, M. et al. The cancer stem cell hypothesis: failures and pitfalls. Neurosurgery 68, 531545 (2011). discussion 545.

Article PubMed Google Scholar

Hewitt, H. B. Studies of the dissemination and quantitative transplantation of a lymphocytic leukaemia of CBA mice. Br. J. Cancer 12, 378401 (1958).

Article CAS PubMed PubMed Central Google Scholar

Park, C. H., Bergsagel, D. E. & McCulloch, E. A. Mouse myeloma tumor stem cells: a primary cell culture assay. J. Natl Cancer Inst. 46, 411422 (1971).

CAS PubMed Google Scholar

Hamburger, A. W. & Salmon, S. E. Primary bioassay of human tumor stem cells. Science 197, 461463 (1977).

Article CAS PubMed Google Scholar

Baccelli, I. & Trumpp, A. The evolving concept of cancer and metastasis stem cells. J. Cell Biol. 198, 281293 (2012).

Article CAS PubMed PubMed Central Google Scholar

Valent, P. et al. Cancer stem cell definitions and terminology: the devil is in the details. Nat. Rev. Cancer 12, 767775 (2012).

Article CAS PubMed Google Scholar

Quintana, E. et al. Phenotypic heterogeneity among tumorigenic melanoma cells from patients that is reversible and not hierarchically organized. Cancer Cell 18, 510523 (2010).

Article CAS PubMed PubMed Central Google Scholar

Taussig, D. C. et al. Anti-CD38 antibody-mediated clearance of human repopulating cells masks the heterogeneity of leukemia-initiating cells. Blood 112, 568575 (2008).

Article CAS PubMed Google Scholar

Ginestier, C. et al. ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell 1, 555567 (2007).

Article CAS PubMed PubMed Central Google Scholar

Grosse-Gehling, P. et al. CD133 as a biomarker for putative cancer stem cells in solid tumours: limitations, problems and challenges. J. Pathol. 229, 355378 (2013).

Article CAS PubMed Google Scholar

Jaksch, M. et al. Cell cycle-dependent variation of a CD133 epitope in human embryonic stem cell, colon cancer, and melanoma cell lines. Cancer Res. 68, 78827886 (2008).

Article CAS PubMed PubMed Central Google Scholar

Singh, S. K. et al. Identification of a cancer stem cell in human brain tumors. Cancer Res. 63, 58215828 (2003).

CAS PubMed Google Scholar

Maitland, N. J. & Collins, A. T. Prostate cancer stem cells: a new target for therapy. J. Clin. Oncol. 26, 28622870 (2008).

Article PubMed Google Scholar

LaBarge, M. A. & Bissell, M. J. Is CD133 a marker of metastatic colon cancer stem cells? J. Clin. Investig. 118, 20212024 (2008).

CAS PubMed PubMed Central Google Scholar

Cheng, J. X., Liu, B. L. & Zhang, X. How powerful is CD133 as a cancer stem cell marker in brain tumors? Cancer Treat Rev. 35, 403408 (2009).

Article CAS PubMed Google Scholar

Wu, Y. & Wu, P. Y. CD133 as a marker for cancer stem cells: progresses and concerns. Stem Cells Dev. 18, 11271134 (2009).

Article CAS PubMed Google Scholar

Glumac, P. M. & LeBeau, A. M. The role of CD133 in cancer: a concise review. Clin. Transl. Med. 7, 18 (2018).

Article PubMed PubMed Central Google Scholar

Meng, X. et al. Both CD133+ and CD133- subpopulations of A549 and H446 cells contain cancer-initiating cells. Cancer Sci. 100, 10401046 (2009).

Article CAS PubMed PubMed Central Google Scholar

Alamgeer, M. et al. Cancer stem cells in lung cancer: evidence and controversies. Respirology 18, 757764 (2013).

Article PubMed PubMed Central Google Scholar

Matsui, W. et al. Clonogenic multiple myeloma progenitors, stem cell properties, and drug resistance. Cancer Res. 68, 190197 (2008).

Article CAS PubMed PubMed Central Google Scholar

Go here to read the rest:
Cancer stem cells: advances in knowledge and implications for cancer therapy - Nature.com

Posted in Stem Cells | Comments Off on Cancer stem cells: advances in knowledge and implications for cancer therapy – Nature.com

Page 41«..1020..40414243..5060..»