Induced Pluripotent Stem Cells (iPS) – UCLA Broad Stem …
By daniellenierenberg
iPSC are derived from skin or blood cells that have been reprogrammed back into an embryonic-like pluripotent state that enables the development of an unlimited source of any type of human cell needed for therapeutic purposes. For example, iPSC can be prodded into becoming beta islet cells to treat diabetes, blood cells to create new blood free of cancer cells for a leukemia patient, or neurons to treat neurological disorders.
In late 2007, a BSCRC team of faculty, Drs. Kathrin Plath, William Lowry, Amander Clark, and April Pyle were among the first in the world to create human iPSC. At that time, science had long understood that tissue specific cells, such as skin cells or blood cells, could only create other like cells. With this groundbreaking discovery, iPSC research has quickly become the foundation for a new regenerative medicine.
Using iPSC technology our faculty have reprogrammed skin cells into active motor neurons, egg and sperm precursors, liver cells, bone precursors, and blood cells. In addition, patients with untreatable diseases such as, ALS, Rett Syndrome, Lesch-Nyhan Disease, and Duchenne's Muscular Dystrophy donate skin cells to BSCRC scientists for iPSC reprogramming research. The generous participation of patients and their families in this research enables BSCRC scientists to study these diseases in the laboratory in the hope of developing new treatment technologies.
View original post here:
Induced Pluripotent Stem Cells (iPS) - UCLA Broad Stem ...
Global Stem Cell Therapy Market 2020: Size, Share, Growth Rate, Revenue and Volume, Key-Players, Top Regions and Forecast Till 2025 – Cole of Duty
By daniellenierenberg
Global Stem Cell Therapy Market report is aimed at highlighting a first-hand documentation of all the best practices in the Stem Cell Therapy industry that subsequently set the growth course active. These vital market oriented details are highly crucial to overcome cut throat competition and all the growth oriented practices typically embraced by frontline players in the Stem Cell Therapy market. Various factors and touch points that the research highlights in the report is a holistic, composite amalgamation of product portfolios of market participants, growth multiplying practices and solutions, sales gateways as well as transaction modes that coherently reflect a favorable growth prospect scenario of the market.
Get sample copy of Stem Cell Therapy Market report @https://www.adroitmarketresearch.com/contacts/request-sample/691
In addition, study report offers an array of opportunities for the players participating in the industry. This ultimately leads into the growth of the global Stem Cell Therapy market. Furthermore, report offers a comprehensive study on market size, revenue, sales, growth factors and risks involved in the growth of the market during the forecast period. The factors which are influencing the growth the market are mentioned in the report as well as the challenges which can hamper the growth of the market over the forecast period.
Full browse the report description and TOC:https://www.adroitmarketresearch.com/industry-reports/stem-cell-therapy-market
The research report encourages the readers to comprehend the importance of quality, shortcomings if any and deep investigation for every member independently by giving the global data of great importance about the market. Consequently, the research report presents the organization profiles and deals investigation of the considerable number of vendors which can assist the customers with taking better choice of the products and services. The end clients of the global Stem Cell Therapy market can be sorted based on size of the endeavour. This research report presents the open doors for the players of the global Stem Cell Therapy market. It additionally offers plans of action which can be taken and market conjectures that would be required.
Global Stem Cell Therapy market is segmented based by type, application and region.
Based on Type, the market has been segmented into:
Based on cell source, the market has been segmented into,
Adipose Tissue-Derived Mesenchymal SCsBone Marrow-Derived Mesenchymal SCsEmbryonic SCsOther Sources
Based on application, the market has been segmented into:
Based on therapeutic application, the market has been segmented into,
Musculoskeletal DisordersWounds & InjuriesCardiovascular DiseasesGastrointestinal DiseasesImmune System DiseasesOther Applications
The company profile section also focusses on companies planning expansions along with mergers & acquisitions, new initiatives, R&D updates and financial updates. But, one of the most important aspects focused in this study is the regional analysis. Region segmentation of markets helps in detailed analysis of the market in terms of business opportunities, revenue generation potential and future predictions of the market. For Stem Cell Therapy market report, the important regions highlighted are North America, South America, Asia, Europe and Middle East. The companies focused on in this report are pioneers in the Stem Cell Therapy market. The uplifting of any region in the global market is dependent upon the market players working in that region.
A qualitative and quantitative analysis of the Stem Cell Therapy market valuations for the expected period is presented to showcase the economic appetency of the global Stem Cell Therapy industry. In addition to this, the global research report comprises significant data regarding the market segmentation which is intended by primary and secondary research methodologies. This research report offers an in-depth analysis of the global Stem Cell Therapy industry with recent and upcoming market trends to offer the impending investment in the Stem Cell Therapy market. The report includes a comprehensive analysis of the industry size database along with the market prediction for the mentioned forecast period. Furthermore, the Stem Cell Therapy market research study offers comprehensive data about the opportunities, key drivers, and restraints with the impact analysis.
Do You Have Any Query Or Specific Requirement? Ask to Our Industry Expert @https://www.adroitmarketresearch.com/contacts/enquiry-before-buying/691
About Us :
Adroit Market Research is an India-based business analytics and consulting company. Our target audience is a wide range of corporations, manufacturing companies, product/technology development institutions and industry associations that require understanding of a markets size, key trends, participants and future outlook of an industry. We intend to become our clients knowledge partner and provide them with valuable market insights to help create opportunities that increase their revenues. We follow a code Explore, Learn and Transform. At our core, we are curious people who love to identify and understand industry patterns, create an insightful study around our findings and churn out money-making roadmaps.
Contact Us :
Ryan JohnsonAccount Manager Global3131 McKinney Ave Ste 600, Dallas,TX 75204, U.S.APhone No.: USA: +1 972-362 -8199 / +91 9665341414
Continue reading here:
Global Stem Cell Therapy Market 2020: Size, Share, Growth Rate, Revenue and Volume, Key-Players, Top Regions and Forecast Till 2025 - Cole of Duty
Bone Marrow Processing System Market to Grow at Robust CAGR in the COVID-19 Lockdown Scenario – 3rd Watch News
By daniellenierenberg
Bone marrow aspiration and trephine biopsy are usually performed on the back of the hipbone, or posterior iliac crest. An aspirate can also be obtained from the sternum (breastbone). For the sternal aspirate, the patient lies on their back, with a pillow under the shoulder to raise the chest. A trephine biopsy should never be performed on the sternum, due to the risk of injury to blood vessels, lungs or the heart.
Request For Report [emailprotected]https://www.trendsmarketresearch.com/report/sample/3184
The need to selectively isolate and concentrate selective cells, such as mononuclear cells, allogeneic cancer cells, T cells and others, is driving the market. Over 30,000 bone marrow transplants occur every year. The explosive growth of stem cells therapies represents the largest growth opportunity for bone marrow processing systems.Europe and North America spearheaded the market as of 2016, by contributing over 74.0% to the overall revenue. Majority of stem cell transplants are conducted in Europe, and it is one of the major factors contributing to the lucrative share in the cell harvesting system market.
Get Complete TOC with Tables and [emailprotected]https://www.trendsmarketresearch.com/report/discount/3184
In 2016, North America dominated the research landscape as more than 54.0% of stem cell clinical trials were conducted in this region. The region also accounts for the second largest number of stem cell transplantation, which is further driving the demand for harvesting in the region.Asia Pacific is anticipated to witness lucrative growth over the forecast period, owing to rising incidence of chronic diseases and increasing demand for stem cell transplantation along with stem cell-based therapy.
<<< Get COVID-19 Report Analysis >>>https://www.trendsmarketresearch.com/report/covid-19-analysis/3184
Japan and China are the biggest markets for harvesting systems in Asia Pacific. Emerging countries such as Mexico, South Korea, and South Africa are also expected to report lucrative growth over the forecast period. Growing investment by government bodies on stem cell-based research and increase in aging population can be attributed to the increasing demand for these therapies in these countries.
Major players operating in the global bone marrow processing systems market are ThermoGenesis (Cesca Therapeutics inc.), RegenMed Systems Inc., MK Alliance Inc., Fresenius Kabi AG, Harvest Technologies (Terumo BCT), Arthrex, Inc. and others
Read more here:
Bone Marrow Processing System Market to Grow at Robust CAGR in the COVID-19 Lockdown Scenario - 3rd Watch News
CRISPR trial shows promising results for sickle cell and thalassaemia – BioNews
By daniellenierenberg
22 June 2020
CRISPRgenome editing has been successfully used to treat three patients with blood disorders in a clinical trial.
Two US patients with beta-thalassaemia and one with sickle cell disease had their bone marrow stem cells edited to produce a different form of haemoglobin, which is normally only found in fetuses and newborns.
'The results [demonstrate] that CRISPR/Cas9 gene editing has the potential to be a curative therapy for severe genetic diseases like sickle cell and beta-thalassaemia,' said Dr Reshma Kewalrami, CEO and President of Vertex, which is running the study jointly with another US pharmaceutical company, CRISPR Therapeutics.
Both sickle cell and beta-thalassaemia are caused by mutations in a gene that produces haemoglobin, the protein in red blood cells that carries oxygen throughout the body. With limited treatment options, patients are often dependent on blood transfusions.
However, the human body is able to make another form of haemoglobin, encoded in a completely separate gene, which is normally only expressed during fetal development and is switched off soon after birth.
In the clinical trial, blood stem cells were removed from the patients and a control gene that turns off the production of fetal haemoglobin was inactivated. Patients were given chemotherapy to remove remaining bone marrow stem cellsbefore they were replaced by the editedcells. The patients were then able to make fetal haemoglobin as adults.
The results of the ongoing trial, presented at the virtual Annual European Hematology Association Congress, reported that two beta-thalassaemia patients were transfusion independent at five and fifteen months after treatment, and the sickle cell patient was free from painful crises at nine months after treatment.
All three patients suffered significant side effects (from which they all recovered), but these were thought to be as a result of the chemotherapy rather than genome editing. Chemotherapy can also have long-term effects including infertility.
It is hoped that this treatment will have long-lasting and durable effects in patients with inherited blood diseases, and early clinical data appear promising. However, patients will need to be followed up throughout their lives to record any changes.
'These highly encouraging early data represent one more step toward delivering on the promise and potential of CRISPR/Cas9 therapies as a new class of potentially transformative medicines to treat serious diseases,' said Dr Samarth Kulkarni, CEO of CRISPR Therapeutics.
See original here:
CRISPR trial shows promising results for sickle cell and thalassaemia - BioNews
Efforts at coronavirus vaccines and treatments abound in the Bay Area – San Francisco Chronicle
By daniellenierenberg
The frenetic search for the miracle that will rid the world of COVID-19 is branching out in a thousand directions, and a large part of the microbial treasure hunt is going on in the Bay Area, where major progress has been made in the 100 days since residents were ordered to shelter in place.
Scientists at universities, laboratories, biotechnology companies and drug manufacturers are combing through blood plasma taken from infected patients for secrets that will help them fight the disease.
The key is likely a super-strength antibody found in some patients. But researchers must first figure out how those antibodies work and how they can be harnessed and used to stop the many health problems associated with COVID-19, particularly acute respiratory distress syndrome, or ARDS, which has killed more people than any other complication connected to the disease.
Other developments showing promise include injections of mesenchymal stem cells, found in bone marrow and umbilical cords, that doctors are studying to battle inflammation caused by ARDS. And a steroid called dexamethasone reduced the number of deaths by halting the overreactive immune responses in seriously ill patients in the United Kingdom.
In all, more than 130 vaccines and 220 treatments are being tested worldwide.
What follows is a list of some of the most promising elixirs, medications and vaccines with ties to the Bay Area:
Monoclonal antibodies / Vir Biotechnology, San Francisco: Scientists at Vir and several institutions, including Stanford and UCSF, are studying monoclonal antibodies, which are clones of coronavirus-fighting antibodies produced by COVID-19 patients.
The idea is to utilize these neutralizing antibodies which bind to the virus crown-like spikes and prevent them from entering and hijacking human cells.
Only about 5% of coronavirus patients have these super-strength antibodies, and those people are believed to be immune to a second attack.
The trick for scientists at Vir is to identify these neutralizing antibodies, harvest, purify and clone them. If they succeed, the resulting monoclones could then be used to inoculate people and it is hoped give them long-term immunity against the coronavirus. The company recently signed a deal with Samsung Biologics, in South Korea, to scale up production of a temporary vaccine in the fall after clinical trials are complete.
Another monoclonal antibody, leronlimab, is being studied in coronavirus clinical trials by its Washington state drugmaker, CytoDyn. The companys chief medical officer is in San Francisco, and the company that does laboratory tests of leronlimab is in San Carlos.
Interferon-lambda / Stanford University: Doctors at Stanford are running a trial to see if interferon-lambda, which is administered by injection, helps patients in the early stages of COVID-19. Interferon-lambda is a manufactured version of a naturally occurring protein that has been used to treat hepatitis. Stanford doctors hope it will boost the immune system response to coronavirus infections.
Dr. Upinder Singh, a Stanford infectious-disease expert, said the trial has enrolled more than 50 patients and is halfway finished. We have noted that patients tolerate the drug very well, she said.
Mesenchymal stem cells / UCSF and UC Davis Medical Center: UCSF Dr. Michael Matthay is leading a study about whether a kind of stem cell found in bone marrow can help patients with ARDS. Matthay hopes that the stem cells can help reduce the inflammation associated with some of ARDS most dire respiratory symptoms, and help patients lungs to recover.
Matthay is aiming to enroll 120 patients in San Francisco, the UC Davis Medical Center in Sacramento and hospitals in a handful of other states. He said the trial, which includes a small number ARDS patients who dont have COVID-19, should have results within a year. So far 17 patients are enrolled in the trial, most of them in San Francisco.
Remdesivir / Gilead Sciences (Foster City): Remdesivir, once conceived as a potential treatment for ebola, was the first drug to show some promise in treating COVID-19 patients. The drug interferes with the process through which the virus replicates itself. A large study led by the federal government generated excitement in late April when officials said hospitalized patients who received remdesivir intravenously recovered faster than those who received a placebo.
A later study looking at dosage showed some benefit for moderately ill COVID-19 patients who received remdesivir for five days, but improvement among those who got it for 10 days was not statistically significant. Gilead, a drug company, recently announced that it will soon launch another clinical trial to see how remdesivir works on 50 pediatric patients, from newborns to teenagers, with moderate to severe COVID-19 symptoms. More than 30 locations in the U.S. and Europe will be involved in the trial, the company said.
Coronavirus crisis: 100 days
Editors note: Its been 100 days since the Bay Area sheltered in place, protecting itself from the coronavirus pandemic. What have we learned in that time? And what does the future hold for the region and its fight against COVID-19? The Chronicle explores the past 100 days and looks to the future in this exclusive report.
Favipiravir / Fujifilm Toyama Chemical (Stanford University): This antiviral drug, developed in 2014 by a subsidiary of the Japanese film company to treat influenza, is undergoing numerous clinical studies worldwide, including a Stanford University trial that began this month. Unlike remdesivir, it can be administered orally, so it can be used to treat patients early in the disease, before hospitalization is necessary.
Stanford epidemiologists want to see if favipiravir, which has shown promising results in other trials, prevents the coronavirus from replicating in human cells, halts the shedding of the virus and reduces the severity of infection. The Stanford study, the only outpatient trial for this drug in the nation, is enrolling 120 people who have been diagnosed with COVID-19 within the past 72 hours. Half of them will get a placebo. People can enroll by emailing treatcovid@stanford.edu.
Colchicine / UCSF (San Francisco and New York): The anti-inflammatory drug commonly used to treat gout flare-ups is being studied in the U.S. by scientists at UCSF and New York University. The drug short-circuits inflammation by decreasing the bodys production of certain proteins, and researchers hope that it will reduce lung complications and prevent deaths from COVID-19. About 6,000 patients are receiving colchicine or a placebo during the clinical trial, dubbed Colcorona, which began in March and is expected to be completed in September.
Selinexor / Kaiser Permanente: Kaiser hospitals in San Francisco, Oakland and Sacramento are studying selinexor, an anticancer drug that blocks a key protein in the cellular machinery for DNA processing, as a potential COVID-19 treatment. The drug has both antiviral and anti-inflammatory properties, and its administered orally, according to Kaisers Dr. Jacek Skarbinski. The trial aims to enroll 250 patients with severe symptoms at Kaiser and other hospitals that are participating nationwide.
VXA-COV2-1 / Vaxart, South San Francisco: The biotechnology company Vaxart is testing this drug to see if it is as effective at controlling COVID-19 as trials have shown it to be against influenza. VXA-COV2-1, the only potential vaccine in pill form, uses the genetic code of the coronavirus to trigger a defensive response in mucous membranes. The hope is that the newly fortified membranes will prevent the virus from entering the body.
Its the only vaccine (candidate) that activates the first line of defense, which is the mucosa, said Andrei Floroiu, Vaxarts chief executive, noting that intravenous vaccines kill the virus after it is inside the body. Our vaccine may prevent you from getting infected at all.
The drug was effective against influenza and norovirus in trials and appears to work on laboratory animals, Floroiu said. He expects trials of VXA-COV2-1 on humans to begin later this summer.
VaxiPatch / Verndari (Napa and UC Davis Medical Center): Napa vaccine company Verndari makes a patented adhesive patch that can deliver a vaccine instead of a shot. Now, the company is trying to make a vaccine for COVID-19 that they can administer through that patch. At UC Davis Medical Center in Sacramento, Verndari researchers are developing a potential vaccine that relies on the coronavirus spike-shaped protein. When injected into a person, the substance would ideally train their body to recognize the virus and fight it off without becoming ill.
A spokeswoman told The Chronicle that the companys preclinical tests have shown early, positive data in developing an immune response. Verndari hopes to move into the next phase of testing in the coming weeks and start clinical trials in humans this year.
If the vaccine is proved effective and safe, patients could receive it through the mail, according to company CEO Dr. Daniel Henderson. The patch would leave a temporary mark on the skin that patients could photograph and send to their doctor as proof they have taken the vaccine, Henderson has said.
Peter Fimrite and J.D. Morris are San Francisco Chronicle staff writers. Email: pfimrite@sfchronicle.com, jd.morris@sfchronicle.com Twitter: @pfimrite, @thejdmorris
Go here to see the original:
Efforts at coronavirus vaccines and treatments abound in the Bay Area - San Francisco Chronicle
Cell Therapy Manufacturing Market: Opportunities Forecast and Value Chain 2020-2030 – Cole of Duty
By daniellenierenberg
Prophecy Market Insights Cell Therapy Manufacturing market research report focuses on the market structure and various factors affecting the growth of the market. The research study encompasses an evaluation of the market, including growth rate, current scenario, and volume inflation prospects, based on DROT and Porters Five Forces analyses. The market study pitches light on the various factors that are projected to impact the overall market dynamics of the Cell Therapy Manufacturing market over the forecast period (2019-2029).
The data and information required in the market report are taken from various sources such as websites, annual reports of the companies, journals, and others and were validated by the industry experts. The facts and data are represented in the Cell Therapy Manufacturing report using diagrams, graphs, pie charts, and other clear representations to enhance the visual representation and easy understanding the facts mentioned in the report.
Get Sample Copy of This Report @ https://www.prophecymarketinsights.com/market_insight/Insight/request-sample/21
The Cell Therapy Manufacturing research study contains 100+ market data Tables, Pie Chat, Graphs & Figures spread through Pages and easy to understand detailed analysis. The predictions mentioned in the market report have been derived using proven research techniques, assumptions and methodologies. This Cell Therapy Manufacturing market report states the overview, historical data along with size, share, growth, demand, and revenue of the global industry.
All the key players mentioned in the Cell Therapy Manufacturing market report are elaborated thoroughly based on R&D developments, distribution channels, industrial penetration, manufacturing processes, and revenue. Also, the report examines, legal policies, and competitive analysis between the leading and emerging and upcoming market trends.
Cell Therapy ManufacturingMarket Key Companies:
harmicell, Merck Group, Dickinson and Company, Thermo Fisher, Lonza Group, Miltenyi Biotec GmBH, Takara Bio Group, STEMCELL Technologies, Cellular Dynamics International, Becton, Osiris Therapeutics, Bio-Rad Laboratories, Inc., Anterogen, MEDIPOST, Holostem Terapie Avanazate, Pluristem Therapeutics, Brammer Bio, CELLforCURE, Gene Therapy Catapult EUFETS, MaSTherCell, PharmaCell, Cognate BioServices and WuXi AppTec.
Segmentation Overview:
Apart from key players analysis provoking business-related decisions that are usually backed by prevalent market conditions, we also do substantial analysis on market segmentation. The report provides an in-depth analysis of the Cell Therapy Manufacturing market segments. It highlights the latest trending segment and major innovations in the market. In addition to this, it states the impact of these segments on the growth of the market.
Request [emailprotected] https://www.prophecymarketinsights.com/market_insight/Insight/request-discount/21
Regional Overview:
The survey report includes a vast investigation of the geographical scene of the Cell Therapy Manufacturing market, which is manifestly arranged into the localities. The report provides an analysis of regional market players operating in the specific market and outcomes related to the target market for more than 20 countries.
Australia, New Zealand, Rest of Asia-Pacific
Key Questions Answered in Report:
Stakeholders Benefit:
About us:
Prophecy Market Insights is specialized market research, analytics, marketing/business strategy, and solutions that offers strategic and tactical support to clients for making well-informed business decisions and to identify and achieve high-value opportunities in the target business area. We also help our clients to address business challenges and provide the best possible solutions to overcome them and transform their business.
Contact Us:
Mr. Alex (Sales Manager)
Prophecy Market Insights
Phone: +1 860 531 2701
Email: [emailprotected]
VISIT MY BLOG:- https://prophecyconsumerelectronics.blogspot.com/
Go here to see the original:
Cell Therapy Manufacturing Market: Opportunities Forecast and Value Chain 2020-2030 - Cole of Duty
Outlook on the Worldwide Regenerative Medicine Industry to 2024 – Rising Global Healthcare Expenditure Presents Opportunities – GlobeNewswire
By daniellenierenberg
Dublin, June 22, 2020 (GLOBE NEWSWIRE) -- The "Global Regenerative Medicine Market: Size & Forecast with Impact Analysis of COVID-19 (2020-2024)" report has been added to ResearchAndMarkets.com's offering.
This report provides an in-depth analysis of the global regenerative medicine market with description of market sizing and growth. The analysis includes market by value, by product, by material and by region. Furthermore, the report also provides detailed product analysis, material analysis and regional analysis.
Moreover, the report also assesses the key opportunities in the market and outlines the factors that are and would be driving the growth of the industry. Growth of the overall global regenerative medicine market has also been forecasted for the years 2020-2024, taking into consideration the previous growth patterns, the growth drivers and the current and future trends.
Region Coverage:
Company Coverage:
Regenerative medicines emphasise on the regeneration or replacement of tissues, cells or organs of the human body to cure the problem caused by disease or injury. The treatment fortifies the human cells to heal up or transplant stem cells into the body to regenerate lost tissues or organs or to recover impaired functionality. There are three types of stem cells that can be used in regenerative medicine: somatic stem cells, embryonic stem cells (ES cells) and induced pluripotent stem cells (iPS cells).
The regenerative medicine also has the capability to treat chronic diseases and conditions, including Alzheimer's, diabetes, Parkinson's, heart disease, osteoporosis, renal failure, spinal cord injuries, etc. Regenerative medicines can be bifurcated into different product type i.e., cell therapy, tissue engineering, gene therapy and small molecules and biologics. In addition, on the basis of material regenerative medicine can be segmented into biologically derived material, synthetic material, genetically engineered materials and pharmaceuticals.
The global regenerative medicine market has surged at a progressive rate over the years and the market is further anticipated to augment during the forecasted years 2020 to 2024. The market would propel owing to numerous growth drivers like growth in geriatric population, rising global healthcare expenditure, increasing diabetic population, escalating number of cancer patients, rising prevalence of cardiovascular disease and surging obese population.
Though, the market faces some challenges which are hindering the growth of the market. Some of the major challenges faced by the industry are: legal obligation and high cost of treatment. Whereas, the market growth would be further supported by various market trends like three dimensional bioprinting , artificial intelligence to advance regenerative medicine, etc.
Key Topics Covered:
1. Executive Summary
2. Introduction2.1 Regenerative Medicine: An Overview2.2 Regeneration in Humans: An Overview2.3 Expansion in Peripheral Industries of Regenerative Medicine2.4 Approval System for Regenerative Medicine Products2.5 Regenerative Medicine Segmentation
3. Global Market Analysis3.1 Global Regenerative Medicine Market: An Analysis3.1.1 Global Regenerative Medicine Market by Value3.1.2 Global Regenerative Medicine Market by Products (Cell Therapy, Tissue Engineering, Gene Therapy and Small Molecules and Biologics)3.1.3 Global Regenerative Medicine Market by Material (Biologically Derived Material, Synthetic Material, Genetically Engineered Materials and Pharmaceuticals)3.1.4 Global Regenerative Medicine Market by Region (North America, Europe, Asia Pacific and ROW)
3.2 Global Regenerative Medicine Market: Product Analysis3.2.1 Global Cell Therapy Regenerative Medicine Market by Value3.2.2 Global Tissue Engineering Regenerative Medicine Market by Value3.2.3 Global Gene Therapy Regenerative Medicine Market by Value3.2.4 Global Small Molecules and Biologics Regenerative Medicine Market by Value
3.3 Global Regenerative Medicine Market: Material Analysis3.3.1 Global Biologically Derived Material Market by Value3.3.2 Global Synthetic Material Market by Value3.3.3 Global Genetically Engineered Materials Market by Value3.3.4 Global Regenerative Medicine Pharmaceuticals Market by Value
4. Regional Market Analysis4.1 North America Regenerative Medicine Market: An Analysis4.2 Europe Regenerative Medicine Market: An Analysis4.3 Asia Pacific Regenerative Medicine Market: An Analysis4.4 ROW Regenerative Medicine Market: An Analysis
5. COVID-195.1 Impact of Covid-195.2 Response of Industry to Covid-195.3 Variation in Organic Traffic5.4 Regional Impact of COVID-19
6. Market Dynamics6.1 Growth Drivers6.1.1 Growth in Geriatric Population6.1.2 Rising Global Healthcare Expenditure6.1.3 Increasing Diabetic Population6.1.4 Escalating Number of Cancer Patients6.1.5 Rising Prevalence of Cardiovascular Disease6.1.6 Surging Obese Population6.2 Challenges6.2.1 Legal Obligation6.2.2 High Cost of Treatment6.3 Market Trends6.3.1 3D Bio-Printing6.3.2 Artificial Intelligence to Advance Regenerative Medicine
7. Competitive Landscape7.1 Global Regenerative Medicine Market Players: A Financial Comparison7.2 Global Regenerative Medicine Market Players' by Research & Development Expenditure
8. Company Profiles8.1 Bristol Myers Squibb (Celgene Corporation)8.1.1 Business Overview8.1.2 Financial Overview8.1.3 Business Strategy8.2 Medtronic Plc8.2.1 Business Overview8.2.2 Financial Overview8.2.3 Business Strategy8.3 Smith+Nephew (Osiris Therapeutics, Inc.)8.3.1 Business Overview8.3.2 Financial Overview8.3.3 Business Strategy8.4 Novartis AG8.4.1 Business Overview8.4.2 Financial Overview8.4.3 Business Strategy
For more information about this report visit https://www.researchandmarkets.com/r/w15smu
Research and Markets also offers Custom Research services providing focused, comprehensive and tailored research.
Read the original:
Outlook on the Worldwide Regenerative Medicine Industry to 2024 - Rising Global Healthcare Expenditure Presents Opportunities - GlobeNewswire
Rapid Unit Sales of Amniotic Fluid Stem Cell Therapy to Account for Incremental Revenues in the Global Market through the COVID 19 Crisis Period -…
By daniellenierenberg
Stem cells are biological cells which have the ability to distinguish into specialized cells, which are capable of cell division through mitosis. Amniotic fluid stem cells are a collective mixture of stem cells obtained from amniotic tissues and fluid. Amniotic fluid is clear, slightly yellowish liquid which surrounds the fetus during pregnancy and is discarded as medical waste during caesarean section deliveries. Amniotic fluid is a source of valuable biological material which includes stem cells which can be potentially used in cell therapy and regenerative therapies. Amniotic fluid stem cells can be developed into a different type of tissues such as cartilage, skin, cardiac nerves, bone, and muscles. Amniotic fluid stem cells are able to find the damaged joint caused by rheumatoid arthritis and differentiate tissues which are damaged. Medical conditions where no drug is able to lessen the symptoms and begin the healing process are the major target for amniotic fluid stem cell therapy. Amniotic fluid stem cells therapy is a solution to those patients who do not want to undergo surgery. Amniotic fluid has a high concentration of stem cells, cytokines, proteins and other important components. Amniotic fluid stem cell therapy is safe and effective treatment which contain growth factor helps to stimulate tissue growth, naturally reduce inflammation. Amniotic fluid also contains hyaluronic acid which acts as a lubricant and promotes cartilage growth.
Get Free Sample Copy With Impact Analysis Of COVID-19 Of Market Report @https://www.persistencemarketresearch.com/samples/23101
With increasing technological advancement in the healthcare, amniotic fluid stem cell therapy has more advantage over the other therapy. Amniotic fluid stem cell therapy eliminates the chances of surgery and organs are regenerated, without causing any damage. These are some of the factors driving the growth of amniotic fluid stem cell therapy market over the forecast period. Increasing prevalence of chronic diseases which can be treated with the amniotic fluid stem cell therapy propel the market growth for amniotic fluid stem cell therapy, globally. Increasing funding by the government in research and development of stem cell therapy may drive the amniotic fluid stem cell therapy market growth. But, high procedure cost, difficulties in collecting the amniotic fluid and lack of reimbursement policies hinder the growth of amniotic fluid stem cell therapy market.
The global amniotic fluid stem cell therapy market is segmented on basis of treatment, application, end user and geography:
Some of the key players operating in global amniotic fluid stem cell therapy market are Stem Shot, Provia Laboratories LLC, Thermo Fisher Scientific Inc. Mesoblast Ltd., Roslin Cells, Regeneus Ltd. etc. among others.
Rapid technological advancement in healthcare, and favorable results of the amniotic fluid stem cells therapy will increase the market for amniotic fluid stem cell therapy over the forecast period. Increasing public-private investment for stem cells in managing disease and improving healthcare infrastructure are expected to propel the growth of the amniotic fluid stem cell therapy market.
You Can Buy This PMR Healthcare Report From Here @https://www.persistencemarketresearch.com/checkout/23101
However, on the basis of geography, global Amniotic Fluid Stem Cell Therapy Market is segmented into six key regionsviz. North America, Latin America, Europe, Asia Pacific Excluding China, China and Middle East & Africa. North America captured the largest shares in global Amniotic Fluid Stem Cell Therapy Market and is projected to continue over the forecast period owing to technological advancement in the healthcare and growing awareness among the population towards the new research and development in the stem cell therapy. Europe is expected to account for the second largest revenue share in the amniotic fluid stem cell therapy market. The Asia Pacific is anticipated to have rapid growth in near future owing to increasing healthcare set up and improving healthcare expenditure. Latin America and the Middle East and Africa account for slow growth in the market of amniotic fluid stem cell therapy due to lack of medical facilities and technical knowledge.
Serena Williams reveals her skincare regimen – HOLA USA
By daniellenierenberg
Serena Williams is an athlete with a busy schedule who has early morning activities. However, she makes sure to go live on Instagram for her Serena Saturdays series where she talks about fashion, beauty and relationships. While Serenas schedule is full of training, the athlete always finds time to practice wellness and is dedicated to show her daughter, Olympia Ohanian how to take care of her skin. In a recent live video, the 38-year-old tennis player revealed her go-to moisturizer and daily skincare practice. One of her most essential beauty product is the eye serum. Eye cream doesnt work unless you put some serum on before, shared Serena.
Though Serena forgot to add serum on her face, she confesses this is an essential step during her skincare regimen. The professional athlete swears by Trilogy Vitamin C Moisturising Lotion which features antioxidants that helps skin prevent damage and recover from free radical exposure. In addition, this moisturizer is known for its radiance-boosting properties which helps the complexion look fresher and brighter. Trilogys formula includes daisy extract and mandarin oil with extra brightening properties such as certified organic Rosehip Seed Oil to help the skin hydrate, replenish and strengthen skins moisture barrier. Aside from using a vegan product, Serena applies Neocutis restorative eye cream after applying eye serum and uses it on her entire face whenever her skin feels extra dry.
Link:
Serena Williams reveals her skincare regimen - HOLA USA
Chitosan Coating of TiO2 Nanotube Arrays for Improved Metformin Releas | IJN – Dove Medical Press
By daniellenierenberg
Amir Hashemi,1 Masoumeh Ezati,2 Javad Mohammadnejad,3 Behzad Houshmand,4 Shahab Faghihi5
1Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran 14395-1561, Iran; 2Tissue Engineering and Biomaterials Research Center, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran 14965/161, Iran; 3Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran 14395-1561, Iran; 4Department of Periodontics, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran 19857-17443, Iran; 5Tissue Engineering and Biomaterials Research Center, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran 14965/161, Iran
Correspondence: Javad Mohammadnejad; Shahab Faghihi Tel +9821 8609-3078Tel/ Fax +98 21 44787386Fax +98(21)88497324Email mohamadnejad@ut.ac.ir; sfaghihi@nigeb.ac.ir
Background: Ineffective integration has been recognized as one of the major causes of early orthopedic failure of titanium-based implants. One strategy to address this problem is to develop modified titanium surfaces that promote osteoblast differentiation. This study explored titanium surfaces modified with TiO2 nanotubes (TiO2 NTs) capable of localized drug delivery into bone and enhanced osteoblast cell differentiation.Materials and Methods: Briefly, TiO2 NTs were subjected to anodic oxidation and loaded with Metformin, a widely used diabetes drug. To create surfaces with sustainable drug-eluting characteristics, TiO2 NTs were spin coated with a thin layer of chitosan. The surfaces were characterized via scanning electron microscopy, atomic force microscopy, and contact angle measurements. The surfaces were then exposed to mesenchymal bone marrow stem cells (MSCs) to evaluate cell adhesion, growth, differentiation, and morphology on the modified surfaces.Results: A noticeable increase in drug release time (3 days vs 20 days) and a decrease in burst release characteristics (85% to 7%) was observed in coated samples as compared to uncoated samples, respectively. Chitosan-coated TiO2 NTs exhibited a considerable enhancement in cell adhesion, proliferation, and genetic expression of type I collagen, and alkaline phosphatase activity as compared to uncoated TiO2 NTs.Conclusion: TiO2 NT surfaces with a chitosan coating are capable of delivering Metformin to a bone site over a sustained period of time with the potential to enhance MSCs cell attachment, proliferation, and differentiation.
Keywords: titania nanotubes, titanium, osteogenic differentiation, anodization, mesenchymal bone marrow stem cells, MSCs
This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License.By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.
See original here:
Chitosan Coating of TiO2 Nanotube Arrays for Improved Metformin Releas | IJN - Dove Medical Press
Orca Bio breaches the surface with $192M for ‘high precision’ cell therapies – FierceBiotech
By daniellenierenberg
Bone marrow transplants can save patients lives by essentially giving them a new immune system to fight off cancer. But they can also cause life-threatening side effects, so their use is relegated to the sickest of patients. Orca Bio wants to change that by taking aim at how these treatments are made.
The Bay Area biotech is coming out of stealth with a $192 million series D round that will propel a pipeline of high precision allogeneic cell therapies and the manufacturing technology behind those treatments. Founded in 2016, Orca Bio zeroed in on manufacturing to make bone marrow transplants safer and more effective.
Theres a bit of a trade-off: You can have precision and a few cells, or you can have lots of cells and sacrifice precision, Orca CEO and co-founder Ivan Dimov, Ph.D., told Fierce Biotech. Most folks out there deal with less precision in order to get the sheer number of cells to treat patients We focused on technology to process extremely large numbers of cells while still having single-cell precision.
Fierce Innovation Awards: Life Sciences Edition 2020
Submit your entry to demonstrate innovative technologies and services that have the potential to make the greatest impact for biotech and pharma companies.
RELATED: AACR: A look at next-gen CAR-T therapies for blood cancers
Orcas proposition is to take donor T cells and stem cells, sort them into their different subtypes and combine them in the right mixture to treat disease.
We dont genetically modify them. But if we now take these cells and build a proprietary mix of them with single-cell precision, we can define the function of what theyre going to do, Dimov said. We can elicit powerful curative effects and control toxicities in a precise way to enhance safety and efficacy in patients that essentially need a whole new blood and immune system.
Dimov likens the processto assembling different kinds of soldiers into the right army unit to give patients so they have a new immune system to seek and destroy cancers while not seeking and destroying the patient themselves and their own tissue.
Because the manufacturing process is quick and uses donor cells, Orcas treatments could eventually reach more patients than CAR-T therapies and other engineered cell therapies can. Some cancer patients may not have enough T cells, or T cells of good enough quality, to turn into a treatment, while others simply do not live long enough for the treatment to be made.
RELATED: BIO: Meet Refuge Biotech, the company developing 'intelligent' cell therapies
The series D, drawn from Lightspeed Ventures, 8VC, DCVC Bio, ND Capital, Mubadala investment Company, Kaiser Foundation Hospitals, Kaiser Permanente Group Trust and IMRF, brings Orcas total raised to nearly $300 million. That haulwill bring its lead program, TRGFT-201, through clinical development. The program is in a phase 1/2 study in patients with blood cancers, while a second program, OGFT-001, is in a phase 1 study, also in blood cancers.
Orcas first two programs are designed for patients with terminal blood cancers, but they could move earlier in the cancer care timeline if they prove to be safer than traditional bone marrow transplants. Beyond cancer, the approach could be applied to a range of genetic disorders of the blood and immune system. The companyhasnt decided where to go next, but Dimov said the approach could be useful in treating autoimmune diseases like Crohns disease or Type 1 diabetes.
Follow this link:
Orca Bio breaches the surface with $192M for 'high precision' cell therapies - FierceBiotech
Magenta Therapeutics and Beam Therapeutics Announce Collaboration to Evaluate Targeted Antibody-Drug Conjugate (ADC) MGTA-117 as Conditioning Regimen…
By daniellenierenberg
CAMBRIDGE, Mass.--(BUSINESS WIRE)--Magenta Therapeutics (Nasdaq: MGTA) and Beam Therapeutics (Nasdaq: BEAM) today announced a non-exclusive research and clinical collaboration agreement to evaluate the potential utility of MGTA-117, Magentas novel targeted ADC for conditioning of patients with sickle cell disease and beta-thalassemia receiving Beams base editing therapies. Beam is pursuing two differentiated base editing approaches to treat hemoglobinopathies: its hereditary persistence of fetal hemoglobin (HPFH) program to precisely and robustly elevate fetal hemoglobin, which could be used in treatments for both sickle cell disease and beta-thalassemia, as well as a novel approach to directly correct the sickle causing point mutation (Makassar).
Conditioning is a critical component necessary to prepare a patients body to receive the edited cells, which carry the corrected gene and must engraft in the patients bone marrow in order to be effective. Todays conditioning regimens rely on nonspecific chemotherapy or radiation, which are associated with significant toxicities. MGTA-117 precisely targets only hematopoietic stem and progenitor cells, sparing immune cells, and has shown high selectivity, potent efficacy, wide safety margins and broad tolerability in non-human primate models. MGTA-117 may be capable of clearing space in bone marrow to support long-term engraftment and rapid recovery in patients.
Beam has demonstrated the ability to edit individual DNA bases in hematopoietic stem cells at high efficiency and with little impact on the viability of edited cells relative to unedited cells using its novel base editing technology. Combining MGTA-117 with Beams HPFH and Makassar base editors could meaningfully advance the treatment of patients with sickle cell disease or beta-thalassemia.
We believe patients will benefit from a more precise process to remove hematopoietic stem cells and prepare them to receive genetic medicines. Magenta has developed targeted ADCs as the preferred modality for our conditioning programs, and we have designed MGTA-117 specifically to optimize it for use with a genetically-modified cell product delivered in a transplant setting, said Jason Gardner, D.Phil., president and chief executive officer, Magenta Therapeutics. Beams next-generation base editing technology complements our next-generation conditioning approach very well, and we are excited to combine these strengths to address the still-significant unmet medical needs of the sickle cell and beta-thalassemia patient communities.
Base editing has the potential to offer lifelong treatment for patients with many diseases, including sickle cell disease and beta-thalassemia. Our novel base editors create precise single base changes in genes without cutting the DNA, enabling durable correction of hematopoietic stem cells with minimal effects on cell viability or genomic integrity, said John Evans, chief executive officer of Beam. Combining the precision of our base editing technology with the more targeted conditioning regimen enabled by MGTA-117 could further improve therapeutic outcomes for patients suffering from these severe diseases. We look forward to partnering with the Magenta team to explore these novel technologies together.
Beam will be responsible for clinical trial costs related to development of Beams base editors when combined with MGTA-117, while Magenta will continue to be responsible for all other development costs of MGTA-117. Magenta will also continue to develop MGTA-117 in other diseases, including blood cancers and genetic diseases. Each company will retain all commercial rights to their respective technologies.
About MGTA-117
MGTA-117, Magentas most advanced conditioning program, is a CD117-targeted antibody engineered for the transplant setting and conjugated to amanitin, a toxin in-licensed from Heidelberg Pharma. It is designed to precisely deplete only hematopoietic stem and progenitor cells and has shown high selectivity, potent efficacy, wide safety margins and broad tolerability in non-human primate models, suggesting that it may be capable of clearing space in bone marrow to support long-term engraftment and rapid recovery in patients. Magenta plans to complete IND-enabling studies this year and initiate clinical studies in 2021. Magenta will continue to develop MGTA-117 in other diseases, including blood cancers and genetic diseases.
About Magenta Therapeutics
Magenta Therapeutics is a clinical-stage biotechnology company developing medicines to bring the curative power of immune system reset through stem cell transplant to more patients with autoimmune diseases, genetic diseases and blood cancers. Magenta is combining leadership in stem cell biology and biotherapeutics development with clinical and regulatory expertise, a unique business model and broad networks in the stem cell transplant world to revolutionize immune reset for more patients. Magenta is based in Cambridge, Mass. For more information, please visit http://www.magentatx.com. Follow Magenta on Twitter: @magentatx.
About Base Editing and Beam TherapeuticsBeam Therapeutics (Nasdaq: BEAM) is a biotechnology company developing precision genetic medicines through the use of base editing. Beams proprietary base editors create precise, predictable and efficient single base changes, at targeted genomic sequences, without making double-stranded breaks in the DNA. This enables a wide range of potential therapeutic editing strategies that Beam is using to advance a diversified portfolio of base editing programs. Beam is a values-driven organization focused on its people, cutting-edge science, and a vision of providing life-long cures to patients suffering from serious diseases. For more information, visit http://www.Beamtx.com.
Magenta Therapeutics Forward-Looking StatementsThis press release may contain forward-looking statements and information within the meaning of The Private Securities Litigation Reform Act of 1995 and other federal securities laws, including, without limitation, statements regarding the research and clinical collaboration agreement between Magenta and Beam, including the timing, progress and success of the collaboration contemplated under the agreement, the successful evaluation of MGTA-117 in conjunction with Beams base-editing therapies under the agreement, the anticipated cost allocation and other commercial terms under the agreement, Magentas strategy and business plan, the future development, manufacture and commercialization between Beam and Magenta as well as statements regarding expectations and plans for the anticipated timing of Magentas clinical trials and regulatory filings and the development of Magentas product candidates and advancement of Magentas preclinical programs. The use of words such as may, will, could, should, expects, intends, plans, anticipates, believes, estimates, predicts, projects, seeks, endeavor, potential, continue or the negative of such words or other similar expressions can be used to identify forward-looking statements. The express or implied forward-looking statements included in this press release are only predictions and are subject to a number of risks, uncertainties and assumptions, including, without limitation, risks set forth under the caption Risk Factors in Magentas most recent Annual Report on Form 10-K filed on March 3, 2020, as updated by Magentas most recent Quarterly Report on Form 10-Q and its other filings with the Securities and Exchange Commission, risks, uncertainties and assumptions regarding the impact of the COVID-19 pandemic to Magentas business, operations, strategy, goals and anticipated timelines, and risks, uncertainties and assumptions inherent in preclinical and clinical studies, including, without limitation, whether results from preclinical studies or earlier clinical studies will be predictive of the results of future trials and the expected timing of submissions for regulatory approval or review by governmental authorities. In light of these risks, uncertainties and assumptions, the forward-looking events and circumstances discussed in this press release may not occur and actual results could differ materially and adversely from those anticipated or implied in the forward-looking statements. You should not rely upon forward-looking statements as predictions of future events. Although Magenta believes that the expectations reflected in the forward-looking statements are reasonable, it cannot guarantee that the future results, levels of activity, performance or events and circumstances reflected in the forward-looking statements will be achieved or occur. Moreover, except as required by law, neither Magenta nor any other person assumes responsibility for the accuracy and completeness of the forward-looking statements included in this press release. Any forward-looking statement included in this press release speaks only as of the date on which it was made. We undertake no obligation to publicly update or revise any forward-looking statement, whether as a result of new information, future events or otherwise, except as required by law.
Beam Forward-Looking Statements
This press release contains forward-looking statements. Investors are cautioned not to place undue reliance on these forward-looking statements, including statements about the timing, progress and success of the collaboration contemplated under the agreement between Beam and Magenta, the successful evaluation of MGTA-117 in conjunction with Beams base-editing therapies under the agreement, the expected timing of filing INDs applications and the therapeutic applications of Beams technology. Each forward-looking statement is subject to risks and uncertainties that could cause actual results to differ materially from those expressed or implied in such statement. Applicable risks and uncertainties include the risks and uncertainties, among other things, regarding: the success in development and potential commercialization of our product candidates; Beams ability to obtain, maintain and enforce patent and other intellectual property protection for our product candidates; whether preclinical testing of our product candidates and preliminary or interim data from preclinical and clinical trials will be predictive of the results or success of ongoing or later clinical trials; that enrollment of clinical trials may take longer than expected; that Beams product candidates will experience manufacturing or supply interruptions or failures; that Beam will be unable to successfully initiate or complete the preclinical and clinical development and eventual commercialization of product candidates; that the development and commercialization of Beams product candidates will take longer or cost more than planned; the impact of COVID-19 on Beams business and the other risks and uncertainties identified under the heading Risk Factors and in Beams Annual Reports on Form 10-K for the year ended December 31, 2019 and in Beams Quarterly Report on Form 10-Q for the quarter ended March 31, 2020, and in any subsequent filings with the Securities and Exchange Commission. These forward-looking statements (except as otherwise noted) speak only as of the date of this press release. Factors or events that could cause Beams actual results to differ may emerge from time to time, and it is not possible for Beam to predict all of them. Beam undertakes no obligation to update any forward-looking statement, whether as a result of new information, future developments or otherwise, except as may be required by applicable law.
Read the original:
Magenta Therapeutics and Beam Therapeutics Announce Collaboration to Evaluate Targeted Antibody-Drug Conjugate (ADC) MGTA-117 as Conditioning Regimen...
Stromal Vascular FractionMarket Estimated to be Driven by Innovation and Industrialization – Personal Injury Bureau UK
By daniellenierenberg
Stromal vascular fraction skin treatment is a type of stem cell therapy based on isolation of adipose tissue during liposuction or lipo-aspiration procedures of patients own body. In stromal vascular fraction treatment isolation of tissue contains fat cells, blood cells, and endothelial cells, as well as a large fraction of adipose-derived mesenchymal stem cells which provides regenerative properties and have positive anti-aging properties. A stromal vascular fraction is considered as a personalized stem cell therapy and effective tropical or injectable treatment.
With increasing age, regenerative and repair properties of skin are less effective due to decrease in stem cell count, and therefore, stromal vascular fraction treatment contains stem cell provides a boost in repair and maintenance mechanism of the skin leaving smooth, healthy, radiant skin. Stromal vascular fraction is a naturally occurring stem cell found in bundles of adipose tissues and are the primary source of growth factors along with macrophages and other cells. Due to the presence of growth factors, the stromal vascular fraction is utilized to decrease inflammation present in many diseases. A stromal vascular fraction is adopted in the treatment of rheumatoid arthritis, joint replacement, osteoarthritis, diabetes, Crohn's disease, and others.
Stromal Vascular Fraction Market: Overview
Get Sample Copy of this report at https://www.xploremr.com/connectus/sample/2376
Stromal vascular fraction is a combination of adipose-derived stromal cells (ADSCs), endothelial cells (ECs), endothelial precursor cells (EPCs), smooth muscle cells, macrophages, pericytes, and pre-adipocytes in the aqueous state. Stromal vascular fraction is advantageous over alternative medical treatments as SVF has the ability to regulate patients own system with the main focus on cell repair and regulation of defective cells. Stromal vascular fraction is a promising field for disease prophylaxis and currently are in clinical trials.
The research report presents a comprehensive assessment of the market and contains thoughtful insights, facts, historical data, and statistically supported and industry-validated market data. It also contains projections using a suitable set of assumptions and methodologies. The research report provides analysis and information according to categories such as market segments, geographies, types, technology and applications.
The report covers exhaustive analysis on: Market Segments Market Dynamics Market Size Supply & Demand Current Trends/Issues/Challenges Competition & Companies involved Technology Value Chain
Stromal Vascular Fraction Market: Segmentation
Request Report Methodology at https://www.xploremr.com/connectus/request-methodology/2376
The globalstromal vascular fraction marketcan be segmented on the basis of type of therapy, end-user, and region.
By Therapy Type SVF Isolation Products Enzymatic Isolation Non-enzymatic Isolation Automated POC Devices SVF Aspirate Purification Products SVF Transfer Products
By End-user Hospitals Specialty Clinics Stem Cell Banks/Laboratories Others
By Application Cosmetic Soft-tissue Orthopedic Others
By Region North America Latin America Europe Asia Pacific (APAC) South Korea Middle East and Africa (MEA)
In its last part, the report offers insights on the key players competing in the global market for stromal vascular fraction. With detailed profiling of each of the key companies active on the competitive landscape, the report provides information about their current financial scenario, revenue share at a global level, development strategies, and future plans for expansion. Strategic collaborations, mergers, and acquisitions have also been considered as a key strategy among a majority of leading companies in the market.
Buy Full Report at https://www.xploremr.com/cart/2376/SL
Read the rest here:
Stromal Vascular FractionMarket Estimated to be Driven by Innovation and Industrialization - Personal Injury Bureau UK
Recent Research Could Restore Vision by Converting Skin Cells into Photoreceptors – Health Writeups
By daniellenierenberg
One of the most complex organs of the human anatomy is the eye. The eye controls the entire visual of an individual, with complex structures and components such as the retina. The retina is a complicated mix of cells and layers that help the eye focus and observe every detail in the field of eyesight.
Any infliction to the retina can result in severe outcomes, potentially leading to retinal diseases. Even the best medications currently like cell therapy can involve a lot of effort and time which the patients cannot give.
However, researchers at the North Texas Eye Research Institute looked into the matter and developed a fast and easier method to rebuild the damaged retina in eye diseases. The method involves a few chemicals that lead to the generation of cells which ultimately restore eyesight.Stem cell therapy
Macular degeneration is a typical reason for loss of eyesight in individuals aged over 60. In this case, the cells which sense light in the retina, primarily known as photoreceptors, begin to deteriorate. Traditionally, doctors have opted for medications for the last surgery to fix this.However, recently, researchers discovered stem cell therapy. This therapy is the method by which loss or degenerated cells are replaced with healthier cells. For the replenishment of these cells, researchers changed the type of specialized cells with the help of specific proteins known as Yamanaka factors.
Reprogramming of specialized cells
This is a big revolution in stem cell therapy. This method can reprogram or restore the generalization of specialized cells such as the heart and immune cells. Basic cells are called pluripotent stem cells. These cells possess the ability to further develop into several types of cells with the inclusion of photoreceptors that are lost in eye diseases.
However, there are rooms for improvements with the struggles in this method. The skin is usually a more typical origin for the reprogramming of cells. The usual time needed is 25 days for the conversion to stem cells. However, further conversion to photoreceptors might take 65-70 extra days, before they are ready to begin cell therapy.
Skipping the reprogramming
With 5 small chemicals, researchers at the North Texas Eye Research Institute have overcome these complications. After publishing their research, they explained that they used these chemicals called small molecule drugs which created photoreceptors straight from skin cells known as fibroblasts, and eliminated the reprogramming step altogether, with no involvement of stem cells.
The 5 chemicals were tested individually and as a combination. Results concluded that the combination showed the most promising results, transforming skin cells into cells that behaved like photoreceptors,
However, the similarity of the photoreceptor-like-cells and the actual photoreceptors, was studied. Hence, they studied the transcriptome of the two, which is an integral part of the cells identity. The results showed sufficient similarity. However, stem cell therapy is a very intricate process with a lot of complications.
The cells have to persist the transplant and change of environment while making the proper connections with the target cells for optimal functionality as a photoreceptor. The real experiment was to test the functionality of these chemically-generated photoreceptors in animal models of eye diseases.
Transplanting the chemically-generated photoreceptors
The transplant was performed in mice with retinal damage. Researchers concluded that almost half of the mice with retinal damage who received the transplant showed pupil reflexes which were similar to mice without retinal damage. This concluded visual response improvement.
The transplant also provided an improved vision, with better pupil reflexes. While these photoreceptors restored vision in the mice with retinal damage, it also helped researchers better understand the cells chemical machinery for the restoration of sight, which lays a foundation for different methods to improve vision.
Read more from the original source:
Recent Research Could Restore Vision by Converting Skin Cells into Photoreceptors - Health Writeups
Discover the latest research in stem cell science during ISSCR 2020 Virtual – 7thSpace Interactive
By daniellenierenberg
Discover the latest research in stem cell science during ISSCR 2020 Virtual
Skokie, IL - Nearly 4,000 members of the global stem cell scientific community will gather virtually 23-27 June to share the latest developments in stem cell research and engage with leaders in the field. ISSCR 2020 Virtual, the annual meeting of the International Society for Stem Cell Research (ISSCR), will feature more than 300 presentations on research areas including clinical innovation and gene editing, stem cells and aging, organogenesis, and machine learning and new computational approaches to research.
What: ISSCR 2020 Virtual, the world's largest meeting dedicated to stem cell research and regenerative medicine
When: 23-27 June, 2020
Where: This is a digital meeting, so join ISSCR 2020 Virtual from anywhere in the world
How: Media may apply for complementary registration for ISSCR 2020 Virtual by going to: https://bit.ly/ISSCRMediaReg. Attendees can register at ISSCR.org.
Credentialed reporters have access to top stem cell researchers, emerging science, and the latest breakthroughs.
Program Highlights:
###
Media are required to register for credentials in order to access ISSCR 2020 Virtual. View ISSCR's credentialing policy.
About the International Society for Stem Cell Research
With nearly 4,000 members from more than 60 countries, the International Society for Stem Cell Research is the preeminent global, cross-disciplinary, science-based organization dedicated to stem cell research and its translation to the clinic. The ISSCR mission is to promote excellence in stem cell science and applications to human health. Additional information about stem cell science is available at A Closer Look at Stem Cells, an initiative of the Society to inform the public about stem cell research and its potential to improve human health.
This story has been published on: 2020-06-19. To contact the author, please use the contact details within the article.
See the original post:
Discover the latest research in stem cell science during ISSCR 2020 Virtual - 7thSpace Interactive
15 Biotech Companies In Houston To Know – Built In
By daniellenierenberg
Houston has played a significant role in boosting the nations biotech industry. While Houston is still a hotspot for energy and oil, the city is steadily becoming a burgeoning life sciences hub. In fact, the city boasted the third fastest-growing biotech community in the nation between 2014 and 2017, according to a CBRE report. Houstons biotech industry is gaining momentum due to an increase in funding as well. According to the Greater Houston Partnership, nearly $180 million in VC funding was allocated to the citys ecosystem of life sciences-related companies in 2019 alone.
Like many startups and tech companies across Houston, the citys life sciences leaders have been tackling some of the worlds most pressing issues. Whether theyre developing oncology drug candidates or advancing genomic medicine through the creation of sequencing technologies, the citys biotech organizations are pulling on decades of research and determination to transform the medical landscape on a global scale. Heres a look at 15 biotech companies in Houston making a major impact on medical research and discovery.
Founded: 2015
Focus: Canine Cancer Treatment
What they do:CAVU Biotherapiesprovides immune-based solutions to treat cancer and autoimmune diseases in dogs. The company offers an immune health monitoring service, which describes a dogs immune system through the use of a blood sample, as well as an autologous prescription product that retrains and expands a dogs T cells to recognize and fight cancer. CAVU Biotherapies ultimate aim is to use its immune-guided medicine to treat horses, cats, andeventually, humans.
Founded: 2006
Focus: Stem Cell Banking + Therapy
What they do: Founded by David Eller and Dr. Stanley Jones, Celltex Therapeutics focuses on developing stem cell therapies for a variety of conditions. The companys stem cell processing and banking methods are designed to ensure the genetic integrity and uniformity of an individuals cells in quantities necessary for therapeutic applications. Using proprietary technology, Celltex Therapeutics enables stem cells to be used for regenerative therapy for conditions like vascular, autoimmune and degenerative diseases.
Founded: 2006
Focus: Cell Therapy
What they do: InGeneron is a clinical stage cell therapy company that specializes in novel, evidence-based regenerative medicine therapies. The companys therapy is designed to repair injured tissue, improve the quality of life for patients and modify the progression of their disease. InGeneron focuses mainly on musculoskeletal indications such as pain management.
Founded: 2006
Focus: Cancer Treatment
What they do: Moleculin Biotech is a pharmaceutical company dedicated to the treatment of highly resistant cancers and viruses. The company develops oncology drug candidates for highly resistant tumors as well as as prodrug to exploit the potential uses of inhibitors of glycolysis. Guided by the aim to provide new hope to cancer patients, Moleculin Biotech focuses on discovering new treatments for acute myeloid leukemia, skin cancer, pancreatic cancer and brain tumors.
Founded: 2001
Focus: Nanomedicine
What they do: Nanospectra Biosciences is spearheading a patient-centric use of nanomedicine for the removal of cancerous tissues. The companys ultra-focal nanoshell technology is designed to thermally destroy solid tumors without damaging adjacent healthy tissue. Nanospectra Biosciences aims to maximize treatment efficacy while minimizing side effects associated with surgery, radiation and traditional focal therapies.
Founded: 2018
Focus: Cell Therapy
What they do: Marker Therapeutics is an immuno-oncology company that focuses on the development of next-generation T cell-based immunotherapies. With the aim of treating hematological malignancies and solid tumor indications, the company uses its own MultiTAA T cell technology, which is based on the selective expansion of non-engineered, tumor-specific T cells. Marker Therapeutics is also working on developing proprietary DNA expression technology that is intended to improve the cellular immune systems ability to recognize and destroy diseased cells.
Founded: 2008
Focus: 3D Cell Culture
What they do: Nano3D Biosciences is dedicated to the development of 3D cell culture solutions. The companys core technology allows them to levitate or bioprint cells, which results in the formation of cultures that are more easily assembled and handled. Nano3D Biosciences products and services are intended for biomedical research, drug discovery, precision medicine, toxicology and regenerative medicine.
Founded: 2017
Focus: Small Molecule Inhibitors
What they do: Tvardi Therapeutics is a clinical-stage biotech company working on a new class of medicines for cancer, chronic inflammation and fibrosis. The company is focusing on the creation of orally delivered, small molecule inhibitors of STAT3, which is a key regulatory protein positioned at the intersection of many disease pathways. Tvardi Therapeutics is dedicated to delivering safe and effective solutions for use in the treatment of numerous diseases.
Founded: 2011
Focus: Targeted Cancer Therapies
What they do: Salarius Pharmaceuticals focuses on developing targeted therapies to treat various types of cancers. The companys lead candidate, Seclidemstat, is intended to treat Ewing sarcoma, a pediatric and young adult bone cancer that currently lacks targeted therapies. Salarius Pharmaceuticals performs clinical trials for the treatment of other advanced solid tumors including prostate, breast and ovarian cancers.
Founded: 2013
Focus: Genomic Medicine
What they do: Founded by Michael Metzker, RedVault Biosciences develops technologies with the aim of advancing genomic medicine. The company is currently working on a variety of projects including the development of sequencing technologies to determine haplotypes and structural variation in complex genomes. RedVault Biosciences is dedicated to identifying technology needs, creating and testing ideas, and transferring deliverables to production and distribution.
Founded: 2010
Focus: DNA Sequencing
What they do: Avance Biosciences focuses on assay development, assay validation and sample testing using next-generation DNA sequencing and other biological methods. The company offers biologics testing, diagnostic assay validation, GMO genomic testing, gene / cell therapy testing, digital and real-time PCR, microbial testing and more. Avance Biosciences aim is to assist its clients in advancing drug development and genomic research.
Founded: 2008
Focus: Bioremediation
What they do: Bionex Technology develops cost-effective, natural solutions for cleaning oil-polluted soil. The companys Super Microbe spill solution is naturally derived from microbes that digest and convert harmful contaminants on the ground and in soil, therefore lowering flammability, suppressing harmful vapors and creating a safer environment for spill responders. Bionex Technology offers a variety of other bioremediation products such as a customizable degreaser and detergent used for cleaning industrial tools.
Founded: 2016
Focus: Stem Cell Research
What they do: Located in nearby Sugar Land, Hope Biosciences is dedicated to developing stem cell-based therapies that are safe, effective and secure. The companys proprietary technology enables patients to make virtually unlimited and identical stem cells from their own tissue. Hope Biosciences offers stem cell banking solutions for both adults and newborns.
Founded: 2013
Focus: Interventional Cardiology
What they do: Saranas has created technology that enables the early detection and monitoring of bleeding complications associated with vascular access procedures. The companys monitoring system checks changes in the blood vessels electrical resistance before monitoring if bleeding has occurred from an unintentionally injured blood vessel. Saranas aims to allow physicians to mitigate downstream consequences by addressing bleeds before they become complications.
Founded: 1984
Focus: Microbiology
What they do: Microbiology Specialists Inc. specializes in microbiology testing, playing a role in microbial investigations and studies. The company also focuses on infectious disease diagnosis, forensic bacteriology and mycology, medical device testing and infection prevention. Microbiology Specialists Inc. is committed to delivering reliable, accurate and cost-effective microbiological results.
Originally posted here:
15 Biotech Companies In Houston To Know - Built In
Nuclear softening expedites interstitial cell migration in fibrous networks and dense connective tissues – Science Advances
By daniellenierenberg
INTRODUCTION
After injury or tissue damage, cells must migrate to the wound site and deposit new tissue to restore function (1). While many tissues provide a permissive environment for such interstitial [three-dimensional (3D)] cell migration (i.e., skin), adult dense connective tissues (such as the knee meniscus, articular cartilage, and tendons) do not support this migratory behavior. Rather, the extracellular matrix (ECM) density and micromechanics increase markedly with tissue maturation (2, 3) and, as a consequence, act as a barrier for cells to reach the wound interface. It follows then that healing of these tissues in adults is poor (4, 5) and that wound interfaces remain susceptible to refailure over the long term due to insufficient repair tissue formation. Similarly, fibrous scaffolds used in repair applications also impede cell infiltration when the scaffolds become too dense (6).
This raises an important conundrum in dense connective tissues and repair scaffolds; while the dense ECM and fibrous scaffold properties are critical for mechanical function, they, at the same time, can compromise cell migration, with endogenous cells locked in place and unable to participate in repair processes. This concept is supported by in vitro studies documenting that, in 3D collagen gels, the migration of mesenchymal lineage cells is substantially attenuated once the gel density and/or stiffness has reached a certain threshold (79). Consistent with this, our recent in vitro models exploring cell invasion into devitalized dense connective tissue (knee meniscus sections) showed reduced cellular invasion in adult tissues compared to less dense fetal tissues (3). The density of collagen in most adult dense connective tissues is 30 to 40 times higher than that used within in vitro collagen gel migration assay systems (2, 3), emphasizing the substantial barrier to migration that the dense ECM plays in these tissues.
To address this ECM impediment to successful healing, we and others have developed strategies to loosen the matrix (via local release of degradative enzymes) in an attempt to expedite repair and/or encourage migration to the wound site (10), with promising results both in vitro and in vivo (10, 11). Despite the potential of this approach, it is cognitively dissonant to disrupt ECM to repair it, and any such therapy would have to consider any adverse consequences on tissue mechanical function.
This led us to consider alternative controllable parameters that might regulate interstitial cell mobility while preserving the essential mechanical functionality of the matrix. It is well established that increasing matrix density decreases the effective pore size within dense connective tissues. The nucleus is the largest (and stiffest) organelle in eukaryotic cells (12), and it must physically deform as a cell passes through constructures that are smaller than its own smallest diameter (9). When artificial pores of decreasing diameter are introduced along an in vitro migration path (e.g., in an in vitro Boyden chamber system), cell motion can be completely arrested (13). If cells are forced to transit through these tight passages, then nuclear rupture and DNA damage can occur (14, 15). Conversely, under conditions where nuclear stiffness is low, as is the case in neutrophils (16) and some particularly invasive cancer cells (17), migration through small pores occurs quite readily.
Given the centrality of the nucleus in migration through small pores, methods to transiently regulate nuclear stiffness or deformability might therefore serve as an effective modulator of interstitial cell migration through dense tissues and scaffolds. Nuclear stiffness is defined by two primary featuresthe density of packing of the genetic material contained within (i.e., the heterochromatin content) and the intermediate filament network that underlies the nuclear envelope (the nuclear lamina, composed principally of the proteins Lamin B and Lamin A/C) (12, 16, 18, 19). Increasing chromatin condensation increases nuclear stiffness, while decreasing Lamin A/C content decreases nuclear stiffness (19, 20). Both increasing the stiffness of the microenvironment in which a cell resides (21) and the mechanical loading history of a cell promotes heterochromatin formation and Lamin A/C accumulation (2224), resulting in stiffer nuclei. Since both matrix stiffening and mechanical loading are features of dense connective tissue maturation, these inputs may drive nuclear mechanoadaptation (25), resulting in endogenous cells with stiff nuclei that are locked in place.
On this basis, the goal of this study was to determine whether nuclear softening could enhance migration through dense connective tissues and repair scaffolds to increase colonization of the wound site and the potential for repair by endogenous cells. We took the approach of transiently decreasing nuclear stiffness in adult meniscus cells through decreasing heterochromatin content [using Trichostatin A (TSA), a histone deacetylase (HDAC) inhibitor] that promotes chromatin relaxation (26) and confirmed the importance of nuclear stiffness by reducing Lamin A/C protein content (using lentiviral-mediated knockdown). Our experimental findings and theoretical models demonstrate that nuclear softening decreases the barriers to interstitial migration through small pores, both in vitro and in vivo, resulting in the improved colonization of dense fibrous networks and transit through native tissue by adult meniscus cells. By addressing the inherent limitations to repair imposed by nuclear mechanoadaptation that accompanies cell differentiation and ECM maturation, this work defines a promising strategy to promote the repair of damaged dense connective tissues in adults.
We first determined whether TSA treatment alters chromatin organization in adult meniscal fibrochondrocytes (MFCs). Super-resolution images of the core histone protein Histone-H2B in MFC nuclei were obtained by stochastic optical reconstruction microscopy (STORM) and revealed a notable organization of Histone-H2B inside MFC nuclei (STORM; Fig. 1A), which could not be observed with conventional microscopy (conventional; Fig. 1A). It has recently been shown that super-resolution images can be segmented at multiple length scales using Voronoi tessellation (27, 28). To segment the H2B super-resolution images, we carried out Voronoi tessellation, used a threshold to remove large polygons corresponding to regions of the nucleus containing sparse localizations, and color-coded the localizations with the same color if their polygons were connected in space and shared at least one edge. This segmentation showed that H2B localizations clustered to form discrete and spatially separated nanodomains in control nuclei [()TSA]. Nuclei treated with TSA, on the other hand, contained smaller domains. These results were quantitatively recapitulated by a decrease in the number of H2B localizations in individual domains and an overall decrease in the area of domains in MFCs treated with TSA [(+)TSA] (Fig. 1, B to D). These results are in line with a more folded chromatin confirmation in ()TSA cells, which opens and decondenses after TSA treatment. These results are also consistent with recent super-resolution analysis, which showed that TSA-treated fibroblasts have small nucleosome nanodomains that are more uniformly distributed in the nuclear space compared to control fibroblasts (29, 30). This decondensation was also confirmed in TSA-treated bovine mesenchymal stem cells (MSCs), where TSA treatment decreased the number and area of H2B nanodomains (fig. S1A). This increased acetylation at H3K9 (Ac-H3K9) was apparent at the nanoscale (fig. S1B) and via conventional fluorescence imaging of the nuclei (fig. S1C). Conversely, there were no significant changes in H3K27me3 with TSA treatment when evaluated using STORM or conventional fluorescent microscopy (fig. S1, D and E).
(A) Representative conventional fluorescent and STORM imaging of Histone-H2B in a control [top; ()TSA] or TSA-treated MFC nucleus [bottom; (+)TSA]. (B) Corresponding Voronoi-based image segmentation, which allows for visualization and quantification of Histone-H2B nanodomains. (C and D) Quantification of the number of H2B localizations per cluster and the cluster area with TSA treatment. The box, line, and dot correspond to the interdecile range (10th to 90th percentile), median, and mean, respectively, Mann-Whitney U test, n 10,584 clusters from five cells. Next to each Voronoi image, higher-magnification zoom-ins of the region inside the squares are shown. (E) TSA treatment for 3 hours decreases chromatin condensation in 4,6-diamidino-2-phenylindole (DAPI)stained nuclei (scale bar, 5 m), and the number of visible edges (left). Quantification of the chromatin condensation parameter (CCP) with TSA treatment [right; *P < 0.05 versus ()TSA, n = ~20]. (F) Schematic showing experimental design to evaluate nuclear deformability and changes in nuclear aspect ratio (NAR = b/a) with cell stretch. (G) Representative DAPI-stained nuclei on scaffolds before and after 15% stretch (left; scale bar, 20 m) and NAR at 3 and 15% stretch (n = 32 to 58 cells, *P < 0.05 versus ()TSA and +P < 0.05 versus 3%). (H) 2D wound closure assay shows no differences in gap filling in the presence or absence of TSA [()TSA; left: scale bar, 200 m; right: P > 0.05, n = 6). (I) Schematic of Boyden chamber chemotaxis assay (left) and migrated cell signal intensity through 3-, 5-, and 8-m-diameter pores, with and without TSA pretreatment [right; n = 5 samples per group, *P < 0.05 versus ()TSA and +P < 0.05 versus 3 m, means SD]. All experiments were carried out at least in triplicate, except for the wound closure assay (which was performed in duplicate). RFU, relative fluorescence units.
In addition, TSA treatment for 3 hours [(+)TSA] also resulted in marked chromatin decondensation in MFCs seeded on aligned (AL) nanofibrous scaffolds that are commonly used for dense connective tissue repair, as evidenced by decreases in the number of visible edges in 4,6-diamidino-2-phenylindole (DAPI)stained nuclei compared to control cells [()TSA] and a reduction (~40%) in the image-based chromatin condensation parameter (CCP) (Fig. 1E).
To assess whether this TSA-mediated chromatin decondensation changed nuclear stiffness and deformability, we stretched MFC-seeded AL scaffolds (from 0 to 15% grip-to-grip strain) and determined the change in nuclear aspect ratio (NAR) (Fig. 1F). Nuclei that were pretreated with TSA [(+)TSA] showed increased nuclear deformation compared to control nuclei [()TSA] (Fig. 1G); however, TSA did not change cell/nuclear morphology (fig. S2, A to C) or cell migration on planar surfaces (Fig. 1H), and only minor changes in focal adhesions were observed (fig. S2, D and E). MFC spread area and traction force generation were also unaffected by TSA treatment when cells were plated on soft substrates (E = 10 kPa) (fig. S2, F to I). These observations suggest that TSA treatment decreases nuclear deformability by chromatin decondensation without changing overall cell migration capacity in 2D culture.
We next assessed the ability of MFCs to migrate through small pores using a commercial transwell migration assay (Fig. 1I). Cells treated with TSA [(+)TSA] (200 ng/ml) showed enhanced migration compared to controls [()TSA] across all pore sizes, including 3-m pores that supported the lowest migration in controls (Fig. 1I). This improved migration with TSA treatment was dose dependent (fig. S3). Together, these data show that while TSA treatment does not change cell morphology, contractility, or planar migration on 2D substrates, chromatin relaxation increases MFC nuclear deformability, which improves cell migration through micron-sized pores.
Having observed increased migration through rigid micron-sized pores with nuclear softening, we next assayed whether TSA treatment would enhance migration through dense fibrillar networks. A custom microfluidic cell migration chamber was designed, consisting of a top reservoir containing basal medium (BM), a bottom reservoir containing BM supplemented with platelet-derived growth factor (PDGF) as a chemoattractant and an interposed nanofibrous poly(-caprolactone) (PCL) layer (labeled with CellTracker Red, ~150-m thickness) (Fig. 2, A and B). With this design, a gradient of soluble factors is presented across the fibrous layer, as evidenced by Trypan blue diffusion over time (Fig. 2C).
(A) Schematic (top) and a top view (bottom) of the PDMS [poly(dimethylsiloxane)]/nanofiber migration chamber. (B) Schematic showing meniscus cells (green) seeded onto fluorescently labeled nanofibers interposed between the top reservoir containing BM and a bottom reservoir containing BM supplemented with PDGF (100 ng/ml) as a chemoattractant. (C) Visual representation of soluble factor gradient in microdevice showing the slow accumulation of trypan blue in the upper chamber as a function of time. (D) Experimental schematic showing meniscus cell (MFC) isolation and seeding onto nanofiber substrates (passage 1, isolated from adult bovine menisci). One day after seeding, TSA or PDGF was added to the top reservoir or the bottom reservoir, respectively, and cells were cultured for additional 2 days. On day 3, scaffolds were imaged by confocal microscopy to determine the degree of cell penetrance into the scaffold. (E) 3D confocal reconstructions of cell (green) migration through AL or non-AL (NAL) nanofibrous networks (AL or NAL; red) with and without TSA treatment. Scale bar, 30 m. (F) Cross-sectional views of cells (green) within nanofibrous substrates (red). Scale bar, 30 m. Quantification of the percentage of infiltrated cells (G) [n = 5 to 8 images, *P < 0.05 versus ()TSA and +P < 0.05 versus AL, means SD] and cell infiltration depth (H) [n = 33 cells, *P < 0.05 versus ()TSA and +P < 0.05 versus AL, means SEM, normalized to the ()TSA/AL group]. Quantification of the percentage of infiltrated cells (I) [n = 5 images, *P < 0.05 versus ()TSA, P < 0.05 versus 0% poly(ethylene oxide) (PEO), and aP < 0.05 versus 25% PEO, means SD] and cell infiltration depth (J) [n = 33 cells, *P < 0.05 versus ()TSA, P < 0.05 versus 0% PEO, and aP < 0.05 versus 25% PEO, means SD] normalized to the control PCL/0% PEO group] as a function of PEO content. All experiments were carried out in triplicate.
MFCs were seeded atop the fibrous layer, and their migration was evaluated as a function of nuclear deformability (TSA) and fiber alignment [AL or non-AL (NAL)]. MFCs were cultured in BM for 1 day for attachment and then were treated for 2 days either with or without TSA (Fig. 2D). Confocal imaging (Fig. 2, E and F, and movie S1, A and B) and scanning electron microscopy (fig. S4A) showed increased MFC invasion into the fibrous networks with TSA treatment [(+)TSA] when compared to untreated MFCs [()TSA]. Without TSA, MFCs remained largely on the surface of the fibers with some cytoplasmic extensions into the fibers (fig. S4B), whereas TSA treatment increased the number of nuclei entering the fiber network (fig. S4C). When quantified, infiltration was higher in the NAL group compared to the AL group (P < 0.05; Fig. 2, G and H), likely due to the increased pore size in the NAL scaffolds (6, 31), and TSA treatment improved migration to similar levels in both NAL and AL groups (P < 0.05; Fig. 2, G and H). As expected, cells in AL scaffolds showed higher aspect ratios and solidity compared to cells on NAL scaffolds, yet TSA treatment did not influence cell morphology (fig. S4D). Nuclei in NAL groups were rounder (lower NAR) than in AL groups, and TSA treatment resulted in more elongated nuclei (higher NAR) in both AL and NAL groups (fig. S4E). While promoting cell invasion, TSA treatment did not result in any change in DNA damage (as assessed by phospho-histone H2AX-positive nuclei; fig. S4F) and slightly reduced cell proliferation at this time point (fig. S4G). Thus, it appears that TSA increased nuclear deformability, resulting in enhanced cell migration into these dense fibrous networks.
To verify that nuclear softening is the primary mechanism for enhanced migration into fibrous networks, we also knocked down Lamin A/C in MFCs before seeding. In previous studies, cells lacking Lamin A/C showed increased nuclear deformability and increased mobility in collagen gels and through small pores in Boyden chambers (13, 32). Consistent with these studies (12, 19, 33), reduction of Lamin A/C protein levels in MFCs and MSCs (fig. S5, A to C) increased nuclear deformability in response to applied stretch (fig. S5D). When MFCs with Lamin A/C knockdown were seeded onto fibrous networks, a greater fraction entered into the scaffold and reached greater infiltration depths (fig. S5, E to G). To further illustrate that nuclear stiffening reduces migration, we cultured MSCs in transforming growth factor3 (TGF-3)containing media for 1 week before seeding onto the fibers. As we reported previously (23), these conditions induce differentiation in MSCs, resulting in stiffer nuclei with increased chromatin condensation and decreased nuclear deformability. Compared to undifferentiated MSCs, these differentiated MSCs were found largely on the scaffold surface (fig. S6, A to D) and had a lower infiltration rate and depth. While many factors change during cell differentiation, these findings also support that a less deformable nucleus is an impediment to interstitial cell migration. Together, these studies support that a stiff nucleus is a limiting factor in the invasion of the small pores of dense fibrous networks.
To investigate the combined role of porosity and nuclear softening on migration, we next fabricated fibrous networks through the combined electrospinning of both PCL and poly(ethylene oxide) (PEO), where PEO acts as a sacrificial fiber fraction to enhance porosity (6, 31). Consistent with our previous findings, cell infiltration percentage and depth progressively increased as a function of increasing PEO content (Fig. 2, I and J). When nuclei were softened with TSA treatment, we observed greater infiltration into low-porosity scaffolds (PEO content, <25%), but no difference in high porosity scaffolds (Fig. 2, I and J). This suggests that increasing nuclear deformability is only beneficial in the context of dense networks, where the nucleus impedes migration.
To better define the relationship between pore size and nuclear stiffness on cellular migration, we developed a computational model to predict the critical force (Fc) required for the nucleus to enter a small channel (Fig. 3). This model was motivated by studies of cellular transmigration through endothelium in the context of cancer invasion, where the surrounding matrix properties (stiffness), endothelium properties (stiffness and pore size), and the cell properties (in particular, the nuclear stiffness) appear to regulate transmigration (34). Here, we considered cell migration into a narrow and long channel to mimic migration into a porous fiber network, where network properties are defined by fiber density (Fig. 3A). When the cell enters the channel, the resistance force encountered by the nucleus increases monotonically as the cell advances, reaching a maximal resistance force (defined as the critical force, Fc). Following this, the nucleus snaps through the opening, leading to a drop in the resistance force, which vanishes after the nucleus fully enters the channel (Fig. 3B and movie S2). Thus, the cells must generate a sufficient force to overcome this critical force to migrate into a channel. As the channel size (rg) becomes smaller and the ECM modulus (EECM) becomes greater, the critical force required for the nucleus to enter the channel increases (Fig. 3C and fig. S7). As this required force increases, it eventually exceeds the force generation capacity of the cell, resulting in a situation where the cell cannot enter the pore.
(A) Schematic showing a nucleus (blue) above a narrow channel representing the small pores in a dense fiber network (orange). The geometric parameters are the radius of the nucleus (rn) and the half width of the channel (rg). The stiffness parameters are the modulus of the nucleus (En) and the fiber network (EECM). The nucleus is treated as a spheroid for simplicity. (B) Simulation of a nucleus moving into and through the channel in the dense fiber network. The normalized resistant force (F/Enrn2) encountered by the nucleus is plotted as a function of the normalized displacement of the nucleus (un/rn). The maximum normalized resistance force is defined as the critical force. (C) The critical force as a function of the normalized ECM modulus (with respect to En) and normalized channel size (with respect to rn). The critical force is larger as the ECM becomes stiffer or the channel becomes smaller. (D) The critical force decreases as the PEO content increases. TSA treatment also decreases the critical force, particularly for dense networks (low PEO content). (E) Normalized NAR after entry into the channel increases as the ECM becomes stiffer or the nucleus becomes softer (both lead to a larger normalized ECM modulus, EECM/En).
To better understand the influence of PEO content (affecting both the channel size and ECM modulus) and dose of TSA (affecting nuclear modulus) on cell migration, we used the normalized critical force data obtained from the model. Our previous work (6) defined the influence of PEO content on matrix mechanical properties and pore size; the effect of TSA on nuclear stiffness has also been measured quantitatively by other groups (26). Using these data, we predicted the critical force at different PEO contents for both TSA-treated and control cells (Fig. 3D). Results from this model showed that critical force decreased monotonically as PEO content increased, given that a higher PEO content results in larger pores (31). This indicates that infiltrated cell numbers should increase as the PEO content increases, consistent with our experimental results. Likewise, since TSA results in a softer nucleus (26), the critical force drops significantly compared to control conditions. This is particularly important at low PEO contents (denser networks), where the critical force for TSA-treated nuclei drops markedly. In networks with larger pores, the difference in critical force between TSA-treated groups vanishes. We included the model to gain, in general, insight into how a change in nuclear deformability (with TSA) might broadly affect cell migration in 3D and chose a simple configuration to gain some initial insight. While this model is simple (i.e., it does not represent the geometry of our fiber networks or native tissue), its predictions were consistent with our experimental findings, where the percentage of infiltrated cells was higher with TSA treatment at 0% PEO but the difference between groups disappeared at 50% PEO (Fig. 2I). The model also predicted that the NAR (after fully embedded in the channel) should increase as the nucleus becomes softer or the ECM becomes stiffer [with both resulting in a larger normalized ECM modulus (Fig. 3E), EECM/En]; this also is consistent with our experimental results showing that the NAR of TSA-treated nuclei within scaffolds was larger than nuclei in the control group.
The above data demonstrate that TSA treatment decreases chromatin condensation for a sufficient period of time to permit migration. However, prolonged exposure to this agent may have deleterious effects on cell phenotype and function. To assess this, we queried how long changes in MFC nuclear condensation persist after TSA withdrawal. MFCs were treated with TSA for 1 day as above, followed by five additional days of culture in fresh BM (Fig. 4A). Consistent with our previous findings, TSA decreased chromatin condensation and CCP after 1 day of treatment (Fig. 4, B and C). Upon removal of TSA, CCP values progressively increased, reaching baseline levels by day 5 (Fig. 4, B and C). A similar finding was noted in H2B localizations and domain area via STORM imaging, where these values returned to baseline levels within 5 days of TSA withdrawal (fig. S8, A to C). Similarly, nuclei in MFCs treated with TSA showed increased deformation compared to control MFC nuclei that were not treated with TSA (Fig. 4D) and increased Ac-H3K9 levels (Fig. 4, E and F), but these values gradually returned to the baseline levels within 5 days with TSA removal (Fig. 4, D to F). Over this same time course, proliferation was decreased in TSA-treated cells but returned to baseline levels within 5 days of TSA withdrawal on both tissue culture plastic (TCP) and on AL nanofibrous scaffolds (fig. S8, D and E). No change in levels of apoptosis (caspase activity) was observed over this time course (fig. 8F). Further, to investigate phenotypic behavior of cells after TSA treatment in the context of tissue repair, we next assayed whether cells exposed to TSA showed alterations in fibrochondrogenic gene expression and collagen production in MFCs. Although the sample size was small in this study, we did not detect a significant change in gene expression for any of the major collagen isoforms or proteoglycans normally produced by meniscus cells (fig. S9A). To further assess this, MFCs were treated with TSA for 1 day, followed by culture in fresh BM or TGF-3 containing chemically defined media (to accelerate collagen production) for an additional 3 days. Collagen produced by these cells and released to the media was not altered by TSA treatment (fig. S9B). Together, these data support that TSA treatment decreases chromatin condensation by increasing acetylation of histones in MFCs but this change is transient and baseline levels are restored gradually after TSA is removed, without alterations in collagen production.
(A) Schematic showing experimental setup; adult MFCs seeded on AL nanofibrous scaffolds were treated with/without TSA in BM for 1 day, followed by culture in fresh BM without TSA for an additional 5 days. (B) Representative DAPI-stained nuclei (top) and corresponding detection of visible edges (bottom) (scale bar, 3 m) and (C) CCP for time points indicated in (A) (red line; BM control at day 0, n = ~20 nuclei, *P < 0.05 versus Ctrl, means SEM). (D) NAR with 3 and 15% of applied stretch (normalized to NAR with 0%, n = 65 ~80 cells, *P < 0.05 versus 3%, +P < 0.05 versus Ctrl, P < 0.05 versus day 0, and aP < 0.05 versus day1, means SEM). (E) Immunostaining for Ac-H3K9 (green) in nuclei (blue) and quantification of mean intensity of the immunostaining (F) (n = ~28 cells, *P < 0.05 versus Ctrl and +P < 0.05 versus day 0, means SEM]. a.u., arbitrary units. All experiments were carried out in triplicate.
Given that transient TSA treatment softened MFC nuclei, resulting in enhanced interstitial cell migration, and did not perturb collagen production in the short term, we next investigated longer-term maturation of a tissue engineered construct with TSA treatment. For this, MFCs were seeded onto AL-PCL/PEO 25% scaffolds and cultured in TGF-3 containing chemically defined media for 4 weeks with/without TSA treatments (once a week for 1 day) as illustrated in Fig. 5A. In controls [()TSA], collagen deposition occurred mostly at the construct border (Fig. 5B), but both deposition and cell distribution were improved with TSA treatment [(+)TSA] (Fig. 5, B and C). Quantification showed that ~50% of cells were located within 50 m of the scaffold edge in controls [()TSA], while TSA treatment [(+)TSA] increased the number of cells deeper within the scaffold (250- to 400-m range; Fig. 5D).
(A) Experimental schematic showing MFCs seeded on PCL/25% PEO nanofibrous scaffolds that were cultured in chemically defined media for 4 weeks with TSA treatment once per week. After 4 weeks, ECM production and cell infiltration with/without TSA treatment were evaluated. Representative cross sections of MFC-laden nanofibrous constructs at week 4 stained for collagen (B) and cell nuclei (C). Scale bar, 100 m. (D) Quantification of MFC infiltration with/without TSA treatment (n = 3 images from three separate samples, *P < 0.05 versus ()TSA, means SEM). Experiments were carried out in duplicate. PSR, Picrosirius Red.
Toward meniscus repair, it is important to evaluate MFC migration through the dense fibrous ECM of meniscus tissue in the context of TSA treatment. For this, adult meniscus explants (, 5 mm) were cultured for ~2 weeks, donor cells in these vital explants were stained with CellTracker, and the explants were placed onto devitalized tissue substrates and cultured for an additional 48 hours, with/without TSA treatment [(/+)TSA] (Fig. 6A). During this 48-hour period, the cells derived from the donor explants adhered to the tissue substrates (Fig. 6B). In control groups [()TSA], cells were found predominantly on the substrate surface, whereas TSA-treated MFCs were found below the substrate surface (Fig. 6, B and C). Quantification showed that both the percent infiltration and the infiltration depth were significantly greater with TSA treatment (Fig. 6D).
(A) Schematic showing processing of vital tissue explants and devitalized tissue sections for invasion assay. Cell migration from the vital tissue and infiltration into the devitalized tissue section were evaluated by confocal microscopy. (B) 3D reconstructions (scale bar, 200 m) and (C) cross-sectional views (scale bar, 50 m) of cells (green) migrating through the devitalized tissue sections (blue), with and without TSA treatment. (D) Quantification of the percentage of infiltrated cells [n = 6 images, *P < 0.05 versus ()TSA, means SD] and cell infiltration depth [n = ~40 cells, *P < 0.05 versus ()TSA, means SEM]. Experiments were carried out in triplicate. (E) Electrospinning schematic showing two independent fiber jets collected simultaneously onto a common rotating mandrel. Discrete fiber populations are composed of PEO containing TSA and PCL. (F) Experimental schematic showing meniscus cell seeding onto nanofiber substrates. One day after seeding, the composite PCL/PEO TSA-releasing (PPT) scaffold was added to the microfluidic chamber reservoir, and cells were cultured for an additional 2 days, followed by confocal imaging. (G) 3D confocal reconstructions of cell (green) migration through AL nanofibrous networks with and without scaffold-mediated TSA delivery (scale bar, 100 m) and quantifications of the percentage of infiltrated cells [n = 5 images, *P < 0.05 versus ()TSA, +P < 0.05 versus (+)TSA, and #P < 0.05 versus 100 ng, means SD; biomolecule loading (mass per scaffold) is based on electrospinning parameters and scaffold mass]. (H) Schematic of repair construct assembly and subcutaneous evaluation in a rat model. (I) Images of DAPI-stained nuclei (blue) at the center of repair constructs after 1 week of subcutaneous implantation, with and without TSA delivery. Dashed lines indicate tissue-scaffold interfaces; dotted lines indicate separation into outer one-third (A), middle (B), and inner one-third (C) sections for quantification. Scale bar, 300 m. (J) Number of cells within each region of the scaffold with and without biomaterial-mediated TSA release (n = 3 samples from three different animals, *P < 0.05 versus PCL/PEO).
Next, we developed an assay to evaluate endogenous cell migration within native tissue. For this, tissue explants (, 6 mm) were excised from adult menisci, and the cells on the periphery of the explants were devitalized using a two-cycle freeze-thaw process (freezing in 20C for 30 min, followed by thawing at room temperature for 30 min, repeated twice on day 2; fig. S10A). This resulted in a ring of dead cells at the periphery of the tissue and a vital core. Processed explants were then treated with TSA for 1 day (day 1) and cultured in fresh media for an additional 3 days (fig. S10A). At the end of culture, living cells along the explant border were quantified. In controls that had not been treated by freeze-thaw (Ctrl), live cells occupied the periphery (fig. S10, B and D). With the two-cycle freeze-thaw process, there was a significant decrease in the number of live cells in this region (fig. S10, B and D), while cells in the center of the explant remained vital (day 2; fig. S10, B and D). With TSA treatment [(+)TSA], the number of vital cells that had migrated from the vital core to the periphery was significantly increased (day 3; fig. S10, C and D).
Last, to demonstrate the clinical potential of these findings, we developed an integrated biomaterial implant system to improve tissue repair in vivo (10, 35) via TSA delivery (Fig. 6E). Here, TSA was released from the PEO fiber fraction of a composite nanofibrous scaffold when this fiber fraction dissolves when placed in an aqueous environment. To first demonstrate bioactivity of the scaffold, we directly included small segments of these TSA-releasing composite scaffolds in the top chamber of the microfluidic migration device to treat seeded MFCs (Fig. 6F). Consistent with findings from soluble delivery, the percentage of infiltrated cells increased with the addition of the TSA-releasing composite scaffold (Fig. 6G): scaffolds releasing ~200 ng of TSA resulted in similar cell migration as direct addition of TSA (200 ng/ml) to the chamber (Fig. 6G). These results show our ability to deliver TSA to the wound site in a controlled fashion. To determine whether these TSA-releasing scaffolds could improve interstitial migration of endogenous meniscus cells in an in vivo setting, we subcutaneously placed meniscal repair constructs in nude rats with empty (PCL/PEO) or TSA-releasing scaffolds (PCL/PEO/TSA) interposed between the cut surfaces and histologically evaluated cellularity of the tissue and implant at 1 week (Fig. 6H). Results showed that interfacial cellularity was markedly higher for repair constructs with the scaffolds releasing ~100 ng of TSA (PCL/PEO/TSA) compared to control scaffolds (PCL/PEO; Fig. 6I), with cells occupying the full thickness of the TSA-releasing scaffold (Fig. 6J). Together, these data indicate that biomaterial-mediated nuclear softening of endogenous meniscus cells increases their capacity for interstitial migration through the tissue and into the scaffold in an in vivo setting.
PCL nanofibrous scaffolds were fabricated via electrospinning as in (6). Briefly, a PCL solution (80 kDa; Shenzhen Bright China Industrial Co. Ltd., China; 14.3% (w/v) in 1:1 tetrahydrofuran and N,N-dimethylformamide) was extruded through a stainless steel needle (2.5 ml/hour, 18-gauge, charged to +13 kV). To form NAL scaffolds, fibers were collected on a mandrel rotating with a surface velocity of <0.5 m/s. For AL scaffolds, fibers were collected at a high surface velocity (~10 m/s) (36). In some studies, to enhance cell infiltration, PCL/PEO (PEO, 200 kDa; Polysciences Inc., Warrington, PA) composite AL fibrous scaffolds were produced by coelectrospinning two fiber fractions onto the same mandrel, as in (6). For this, solutions of PCL (14.3%, w/v) and PEO (10%, w/v, in 90% ethanol) were electrospun simultaneously onto a centrally located mandrel (~10 m/s, 2.5 ml/hour). Resulting composite scaffolds were produced with PEO content of 0, 25, and 50% by scaffold dry mass. To visualize fibers, CellTracker Red (0.0005%, w/v) was mixed into the PCL solutions before electrospinning. Scaffolds were hydrated and sterilized in ethanol (100, 70, 50, and 30%; 30 min per step) and incubated in a fibronectin (20 g/ml) solution overnight to enhance initial cell attachment. TSA-releasing scaffolds contained a semipermanent (very slow degrading) fiber population (PCL) and a transient (water soluble) fiber population (PEO). The PEO fibers released TSA as they dissolve. To form this fiber fraction, TSA was added to the PEO solution (1% wt/vol) 2 days before spinning. PCL (10 ml) and PEO/TSA (10 ml) solutions were loaded into individual syringes and electrospun simultaneously by coelectrospinning onto a common centrally located mandrel, as above. Estimates of TSA content (mass per scaffold) were based on electrospinning parameters and the mass of each fiber fraction (Fig. 6E).
MFCs were isolated from the outer zone of adult bovine (20 to 30 months; Animal Technologies Inc.) or porcine menisci (6 to 9 months; Yucatan, Sinclair BioResources). For this, meniscal tissue segments were minced into ~1-mm3 cubes and placed onto TCP and incubated at 37C in a BM consisting of Dulbeccos modified Eagles medium (DMEM) with 10% fetal bovine serum and 1% penicillin/streptomycin/fungizone (PSF). Cells gradually emerged from the small tissue segments over 2 weeks, after which the remaining tissue was removed and the cells were passaged one time before use. MSCs were isolated from juvenile bovine bone marrow as in (37) and expanded in BM. To induce MSC fibrochondrogenesis, passage 1 MSCs were seeded on AL PCL scaffolds and cultured in a chemically defined serum free medium consisting of high glucose DMEM with 1 PSF, 0.1 M dexamethasone, ascorbate 2-phosphate (50 g/ml), l-proline (40 g/ml), sodium pyruvate (100 g/ml), insulin (6.25 g/ml), transferrin (6.25 g/ml), selenous acid (6.25 ng/ml), bovine serum albumin (BSA; 1.25 mg/ml), and linoleic acid (5.35 g/ml) (Life Technologies, NY, USA). This base medium (Ctrl) was further supplemented with TGF-3 (10 ng/ml) to induce differentiation (Ctrl/Diff, R&D Systems, Minneapolis, MN). Cell-seeded constructs were cultured in this medium for up to 7 days.
MFCs or MSCs were plated into eight-well Lab-Tek 1 cover glass chambers (Nunc), followed by preculture in BM for 2 days. At this time point, cells were treated with TSA for 3 hours, followed by fixation in methanol-ethanol (1:1) at 20C for 6 min. After a 1-hour incubation in blocking buffer containing 10 weight % BSA (Sigma-Aldrich) in phosphate-buffered saline (PBS), samples were incubated overnight with anti-H2B (1:50; abcam1790, Abcam), anti-H3K4me4 (1:100; MA5-11199, Thermo Fisher Scientific), or anti-H3K27me3 (1:100; PA5-31817, Thermo Fisher Scientific) at 4C. Next, samples were washed and incubated for 40 min with secondary antibodies custom labeled with activator-reporter dye pairs (Alexa Fluor 405Alexa Fluor 647, Invitrogen) for STORM imaging (29, 38). All imaging experiments were carried out with a commercial STORM microscope system from Nikon Instruments (N-STORM). For imaging, the 647-nm laser was used to excite the reporter dye (Alexa Fluor 647, Invitrogen) to switch it to the dark state. Next, a 405-nm laser was used to reactivate the Alexa Fluor 647 in an activator dye (Alexa Fluor 405)facilitated manner. An imaging cycle was used in which one frame belonging to the activating light pulse (405 nm) was alternated with three frames belonging to the imaging light pulse (647 nm). Imaging was carried out in a previously described imaging buffer [Cysteamine (#30070-50G, Sigma-Aldrich), GLOX solution: 1 glucose oxidase (0.5 mg/ml), 1 catalase (40 mg/ml) (all from Sigma-Aldrich), and 10% glucose in PBS] (39). STORM images were analyzed and rendered using custom-written software (Insight3, gift of B. Huang, University of California, San Francisco, USA) as previously described (39). For quantitative analysis, a previously described method was adapted that segments super-resolution images based on Voronoi tessellation of the fluorophore localizations (27, 28). Voronoi tessellation of a STORM image assigns a Voronoi polygon to each localization, such that the polygon area is inversely proportional to the local localization density (40). The spatial distribution of localizations is represented by a set of Voronoi polygons such that smaller polygon areas correspond to regions of higher density. Domains were segmented by grouping adjacent Voronoi polygons with areas less than a selected threshold, and imposing a minimum of three localizations per domain criteria generates the final segmented dataset.
MFCs (P1) were seeded onto AL PCL (0% PEO) scaffolds in BM for 2 days. To induce chromatin decondensation, TSA, a HDAC inhibitor (26) was added to the media for 3 hours. Chromatin condensation state and nuclear deformability were evaluated 3 hours after TSA treatment. For chromatin condensation analysis, constructs were fixed in 4% paraformaldehyde for 30 min at 37C, followed by PBS washing and permeabilization (with 0.05% Triton X-100 in PBS supplemented with 320 mM sucrose and 6 mM magnesium chloride). Nuclei were visualized by DAPI (ProLong Gold Antifade Reagent with DAPI, P36935, Molecular Probes, Grand Island, NY) and imaged at their mid-section using a confocal microscope (Leica TCS SP8, Leica Microsystems Inc., IL). Edge density in individual nuclei was measured using a Sobel edge detection algorithm in MATLAB to calculate the CCP as described in (24).
To assess nuclear deformability, the NAR (NAR = a/b) was evaluated before (0%) and after 9 and 15% grip-to-grip static deformation of constructs. Nuclear shape was captured on an inverted fluorescent microscope (Nikon T30, Nikon Instruments, Melville, NY) equipped with a charge-coupled device camera at each deformation level. NAR was calculated using a custom MATLAB code. Changes in NAR were tracked for individual MSC nuclei at each strain step as in (41).
To assess MFC migration on 2D substrates, a scratch assay was performed with or without TSA treatment. For this, passage 1 MFCs were plated into a six-well tissue culture dish (2 105 cells per well) and cultured to confluence (for 2 to 3 days). Confluent monolayers were then scratched with a 2.5-l pipette tip, and cell debris was removed via PBS washing. Images were taken using an inverted microscope at regular intervals and wound closure computed using ImageJ.
In addition, as an initial assessment of MFC migration, 96-well transwell migration assay kits (Chemicon QCM 96-well Migration Assay; membrane pore size, 3, 5, or 8 m) were used to assess cell migration. Briefly, human recombinant PDGF-AB (100 ng/ml in 150 l of BM; Prospect Bio) was added to the bottom chamber, and passage 1 MFCs (50,000 cells per well) were seeded into the top chamber. Cells were allowed to migrate for 18 hours at 37C with/without TSA treatment. In some studies, different dosages of TSA (0 to 800 nM) were applied (at a pore size of 5 m).
To assess initial cell migration through dense nanofiber networks, a custompoly(dimethylsiloxane) (PDMS) migration assay chamber was implemented (Fig. 2A). Top and bottom pieces containing holes (top, 6, 7, 6 mm in diameter; bottom, 6, 5, 6 mm in diameter) and a channel (bottom, 2 mm in width and 20 mm in length) were designed via SOLIDWORKS software for 3D printed templates (Acura SL 5530, Protolabs), and these were cast from the templates with PDMS (Sylgard 184, Dow Corning). To assemble the multilayered chamber, bottom PDMS pieces, the periphery of PCL electrospun fiber networks, and top PDMS pieces were coated with uncured PDMS base and curing agent mixture (10:1 ratio) and placed on cover glasses sequentially. For firm adhesion of each layer, chambers were incubated at 40C overnight. The final device consisted of a top reservoir containing BM and a bottom reservoir containing BM + PDGF (100 ng/ml) as a chemoattractant (Fig. 2A). To simulate chemoattactant diffusion from bottom to top reservoirs, trypan blue 0.4% solution (MP Biomedicals) was introduced to one of the side holes to fill the bottom reservoir, and the central top reservoir was filled with PBS. Cell migration chambers were kept in incubator (37C, 5% CO2), and images were obtained at regular intervals (Fig. 2D).
Fluorescently labeled (CellTracker Red) AL or NAL nanofibrous PCL scaffolds (thickness, ~150 m) were interposed between the reservoirs, and MFCs (2000 cells, passage 1) were seeded onto the top of each scaffold, followed by 1 day before culture in BM. Cells in chambers were cultured in BM with/without TSA for an additional 2 days. At the end of 3 days, cells were fixed and visualized by actin/DAPI staining. Confocal z-stacks were obtained at 40 magnification, and maximum z-stack projections were used to assess cellular morphology (cell/nuclear aspect ratio, area, circularity, and solidity). The percentage of infiltrated cells was quantified from confocal z stacks, with cells located beneath fibers categorized as infiltrated (fig. S3C) and the infiltration depth measured on cross-sectional images using ImageJ. For scanning electron microscopy imaging, additional samples were fixed and dehydrated in ethanol (30, 50, 70, and 100%, 60 min per step) and then hexamethyldisilane for terminal dehydration under vacuum.
Details on the model have been described previously (34). Briefly, to understand the influence of both intracellular and extracellular cues on cell migration through the fibrous ECM, we considered a model in which a cell with a spherical nucleus of radius rn is invading ECM through a deformable gap (with radius rg) smaller than the diameter of the nucleus (Fig. 3A). For simplicity, the nucleus is modeled by a spheroid and treated as a compressible neo-Hookean hyperelastic material to capture the mechanical response. An infinitely long small channel is created in the ECM to mimic the path a cell would migrate through in the migration assay. A neo-Hookean hyperelastic material was used to capture the ECM mechanical properties. The model parameters are shown in Table 1.
To assess how fast the TSA-mediated MFC chromatin organization and deformability was restored after TSA removal, MFCs seeded on AL scaffolds were treated with TSA for 1 day, followed by additional culture for 5 days in fresh BM (Fig. 4A). At each time point, the CCP and nuclear deformability were evaluated as described above. In addition, Ac-H3 levels in MFC nuclei were assessed by immunostaining with an Ac-H3K9 monoclonal antibody (MA5-11195, Thermo Fisher Scientific; 1:400, overnight at 4C). All images were collected using a confocal microscope (Leica TCS SP8, Leica Microsystems Inc., IL) at 63 magnification, with staining intensity quantified using ImageJ.
For long-term evaluation of matrix production after TSA treatment, MFCs were seeded on PCL/PEO 25% AL nanofibrous scaffolds (P1, 105 cells, 1 cm by 1 cm by 0.1 cm) and were cultured in TGF-3 containing chondrogenic media for 4 weeks. TSA was applied once each week for 24 hours. After 4 weeks, constructs were fixed with 4% paraformaldehyde and embedded in CryoPrep frozen section embedding medium [optimal cutting temperature (OCT) compound, Thermo Fisher Scientific, Pittsburgh, PA]. Using a cryostat microtome (Microm HM-500 M Cryostat, Ramsey, MN), constructs were sectioned to 8 m in thickness through their depth and stained with Picrosirius Red and DAPI to visualize collagen and nuclei, respectively. Stained sections were visualized and imaged by brightfield and fluorescent microscopy (Nikon Eclipse TS 100, Melville, NY). To quantify cell infiltration in the scaffolds, the number of migrated cells as a function of scaffold depth was determined for each experimental group (n = 3 scaffolds per group) using ImageJ.
To isolate fresh MFCs, cylindrical tissue explants (6 mm in diameter and 3 mm in height) were excised using biopsy punches from the middle zone of the meniscus, and these explants incubated in BM for ~2 weeks to allow cells to occupy the periphery. To fabricate devitalized tissue substrates, additional cylindrical tissue explants (8 mm in diameter) were embedded in OCT sectioning medium (Sakura Finetek, Torrance, CA) and axially cut (to ~50 m in thickness) using a cryostat microtome. These devitalized sections were placed onto positively charged glass slides and stored at 20C until use. After ~2 weeks of in vitro culture, the living explants were incubated in 5-chloromethylfluorescein diacetate (5 g/ml) (CellTracker Green, Thermo Fisher Scientific, Waltham, MA) in serum-free media (DMEM with 1% PSF) for 1 hour to fluorescently label cells in the explants. The explants were placed atop tissue substrates to allow for cell egress onto and invasion into the sections, and slides with explants were incubated at 37C with/without TSA treatment in BM for 2 days, at which point maximum z-stack projections were acquired using a confocal microscope (Leica TCS SP8, Leica Microsystems Inc., IL). Cell infiltration depth was measured as the distance between the apical tissue surface and the basal cell surface using a custom MATLAB code (3), and the total number of cells and the number of migrated cells (those entirely embedded within the tissue) were counted (n = 3 per group) using ImageJ.
In addition, to observe endogenous meniscus cell migration in the native ECM, a tissue-based migration assay was developed. Cylindrical meniscus tissue explants (6 mm in diameter and ~6 mm in height) were excised from the middle zone of adult menisci. To kill the cells on the border of the tissue, explants were frozen at 20C for 30 min and then thawed at room temperature for 30 min; this process was repeated twice (two-cycle) (day 2; fig. S10A). After devitalizing the periphery, explants were cultured in BM for 1 day, and TSA was added for 1 day (day 1; fig. S10A). After TSA treatment, explants were washed with PBS (day 0; fig. S10A), followed by culture in fresh BM for an additional 3 days. At day 3, LIVE/DEAD staining was performed, and explants cross sections were imaged (day 3; fig. S10A). Images were acquired from eight regions distributed evenly around the boundary (Leica TCS SP8, Leica Microsystems Inc., IL). The number of live cells located within 1 mm of the boundary was determined using ImageJ.
To evaluate the impact of biomaterial-mediated TSA delivery on endogenous meniscus cell migration in an in vivo setting, a nude rat xenotransplant model was used, as in (10). All animal procedures were approved by the Animal Care and Use Committee of the Corporal Michael Crescenz VA Medical Center. Before subcutaneous implantation, horizontal defects were created in adult bovine meniscal explants (8 mm in diameter and 4 mm in height, n = 3 donors; Fig. 6H). Electrospun PCL/PEO scaffolds with/without TSA were prepared (6 mm in diameter with a 2-mm-diameter central fenestration). Control PCL/PEO scaffolds or scaffolds releasing TSA (PCL/PEO/TSA, ~100 ng) were placed into the defect, which was closed with absorbable sutures. The repair construct was implanted subcutaneously into the dorsum of male athymic nude rats (n = 3, Hsd:RH-Foxn1rnu, 8 to 10 weeks old, ~300 g, Harlan) (Fig. 6H) (10). At 1 week, rats were euthanized, and constructs were removed from the subcutaneous space. Samples were fixed with para-formaldehyde and embedded in OCT sectioning medium (Sakura Finetek, Torrance, CA), sectioned to 8 m in thickness, stained with DAPI for cell nuclei, and imaged using a fluorescence microscope. Cell number in the center and edges of the implanted scaffold were determined using ImageJ.
Statistical analysis was performed using Student t tests or analysis of variance (ANOVA) with Tukeys honestly significantly different post hoc tests (SYSTAT v.10.2, Point Richmond, CA). For datasets that were not normally distributed, nonparametric Mann-Whitney or Kruskal-Wallis tests were performed, followed by post hoc testing with Dunns correction using GraphPad Prism version 6 (GraphPad Software Inc., La Jolla, CA, USA). Results are expressed as the means SEM or SD, as indicated in the figure legends. Differences were considered statistically significant at P < 0.05.
Acknowledgments: We acknowledge S. Gullbrand, D. H. Kim, and E. Henning for technical support. Funding: This work was supported by the NIH (R01 AR056624), the Department of Veterans Affairs (I01 RX000174), the NSF Science and Technology Center for Engineering Mechanobiology (CMMI-1548571), and the Penn Center for Musculoskeletal Disorders (P30 AR069619). Author contributions: S.-J.H., K.H.S., S.T., X.C., A.P.P., B.N.S., F.Q., V.B.S., M.L., J.A.B., and R.L.M. designed the studies. S.-J.H., K.H.S., S.T., X.C., A.P.P., and B.N.S. performed the experiments. S.-J.H., K.H.S., S.T., X.C., A.P.P., B.N.S., F.Q., V.B.S., M.L., J.A.B., and R.L.M. analyzed and interpreted the data. S.-J.H., S.T., X.C., V.B.S., M.L., J.A.B., and R.L.M. drafted the manuscript, and all authors edited the final submission. Competing interests: The authors declare that they have no competing interests. Data and materials availability: All data needed to evaluate the conclusions in the paper are present in the paper and/or the Supplementary Materials. Additional data related to this paper may be requested from the authors.
See the original post here:
Nuclear softening expedites interstitial cell migration in fibrous networks and dense connective tissues - Science Advances
For babies born with a rare immune deficiency, a unique new test to better target care – Yahoo India News
By daniellenierenberg
Montreal [Canada], June 18 (ANI): A new test developed at CHU Sainte-Justine in Montreal will enable better management of patients with severe combined immunodeficiency (SCID).
The results of the study were presented in the medical journal Blood Advances published by the American Society of Hematology.
Routine neonatal screening, although not yet available in Quebec, has led to an increase in the incidence of patients diagnosed with SCID in North America in recent years.
This syndrome, a group of rare hereditary genetic disorders, is characterized by a total absence of immune system function, including an absence of T-lymphocytes, the white blood cells that play a crucial role in the body's immune defence.
Without appropriate treatment, the disorder is fatal during the first months of life in the majority of cases.
Many of the genes involved in SCID have been identified, but clinicians sometimes come across patients who do not have any identified genetic abnormalities.
"It's very frustrating. In about seven per cent of patients, we can't provide optimal care because we don't know the genetic cause," said Dr. Elie Haddad, a pediatric immunologist at CHU Sainte-Justine and expert in the field of SCID.
"Depending on the nature of the mutated gene, there are two treatments for SCID: either a bone marrow transplantation or a thymus transplantation. We still need to be able to identify the type of disease in order to choose the correct treatment option," added Haddad.
The gene involved can either disrupt hematopoietic stem cells in the bone marrow that consequently cannot naturally become T-cells, or it can affect the function of the thymus. The thymus is an organ in which immature white blood cells from the bone marrow 'learn' to become T-cells.
When doctors are unable to identify the real cause of the disorder, they usually turn to bone marrow transplantation. They do so for two reasons: first, transplants are easier to perform, and second, among the known genes, more are responsible for a dysfunction of the hematopoietic cells than for a malfunction of the thymus.
However, knowing the origin of the disease is critical, because if it's the thymus that's not working properly, then the bone marrow transplant will have no effect, and vice versa.
"Given this clinical need, our goal was to create a functional test by taking a very small volume of peripheral blood rather than a bone marrow sample, which is a more complex process to perform in babies and more invasive than a simple blood test," said Panojot Bifsha, first author of the study.
In the laboratory, a very small number of stem cells is isolated from patients using a limited amount of blood (3 to 5 mL). A test with a 3D culture that mimics the function of a human thymus is used to test this small number of cells, and a response is obtained in less than five weeks. If the results are normal, thymus transplantation is recommended, but if they are abnormal, then a bone marrow transplant is preferred.
"Our 3D culture system is unique because it allows us to test a very small number of stem cells circulating in the blood and get a relatively quick response. We received blood samples from all over North America, which allowed us to validate our method.
A similar study conducted with bone marrow samples at the U.S. National Institutes of Health (NIH) produced similar results, proving the reliability of the test developed at CHU Sainte-Justine from a blood sample. The U.S. study was also published today in Blood Advances.
As Quebec's hub of care and research for children with rare or serious diseases, CHU Sainte-Justine strives to stay one step ahead in research niches for which it is famous, such as the genetics of rare diseases and innovative treatments in precision medicine.
Additional studies will be required to further validate the latest test and allow it to be used on more patients.(ANI)
Read the original post:
For babies born with a rare immune deficiency, a unique new test to better target care - Yahoo India News
Rheumatoid Arthritis Stem Cell Therapy Market Future Innovation Strategies, Growth & Profit Analysis, Forecast by 2028 – The Cloud Tribune
By daniellenierenberg
The global Rheumatoid Arthritis Stem Cell Therapy market study presents an all in all compilation of the historical, current and future outlook of the market as well as the factors responsible for such a growth. With SWOT analysis, the business study highlights the strengths, weaknesses, opportunities and threats of each Rheumatoid Arthritis Stem Cell Therapy market player in a comprehensive way. Further, the Rheumatoid Arthritis Stem Cell Therapy market report emphasizes the adoption pattern of the Rheumatoid Arthritis Stem Cell Therapy across various industries.Request Sample Reporthttps://www.factmr.com/connectus/sample?flag=S&rep_id=1001The Rheumatoid Arthritis Stem Cell Therapy market report highlights the following players:The global market for rheumatoid arthritis stem cell therapy is highly fragmented. Examples of some of the key players operating in the global rheumatoid arthritis stem cell therapy market include Mesoblast Ltd., Roslin Cells, Regeneus Ltd, ReNeuron Group plc, International Stem Cell Corporation, TiGenix and others.
The Rheumatoid Arthritis Stem Cell Therapy market report examines the operating pattern of each player new product launches, partnerships, and acquisitions has been examined in detail.Important regions covered in the Rheumatoid Arthritis Stem Cell Therapy market report include:
North America (U.S., Canada)Latin America (Mexico, Brazil)Western Europe (Germany, Italy, U.K., Spain, France, Nordic countries, BENELUX)Eastern Europe (Russia, Poland, Rest Of Eastern Europe)Asia Pacific Excluding Japan (China, India, Australia & New Zealand)JapanMiddle East and Africa (GCC, S. Africa, Rest Of MEA)
The Rheumatoid Arthritis Stem Cell Therapy market report takes into consideration the following segments by treatment type:
Allogeneic Mesenchymal stem cellsBone marrow TransplantAdipose Tissue Stem Cells
The Rheumatoid Arthritis Stem Cell Therapy market report contain the following distribution channel:
HospitalsAmbulatory Surgical CentersSpecialty ClinicsHave Any Query? Ask our Industry Experts-https://www.factmr.com/connectus/sample?flag=AE&rep_id=1001
Buy the report at a discounted rate!!! Exclusive offer!!!
The Rheumatoid Arthritis Stem Cell Therapy market report offers a plethora of insights which include:
Changing consumption pattern among individuals globally.Historical and future progress of the global Rheumatoid Arthritis Stem Cell Therapy market.Region-wise and country-wise segmentation of the Rheumatoid Arthritis Stem Cell Therapy market to understand the revenue, and growth lookout in these areas.Accurate Year-on-Year growth of the global Rheumatoid Arthritis Stem Cell Therapy market.Important trends, including proprietary technologies, ecological conservation, and globalization affecting the global Rheumatoid Arthritis Stem Cell Therapy market.
The Rheumatoid Arthritis Stem Cell Therapy market report answers important questions which include:
Which regulatory authorities have granted approval to the application of Rheumatoid Arthritis Stem Cell Therapy in Health industry?How will the global Rheumatoid Arthritis Stem Cell Therapy market grow over the forecast period?Which end use industry is set to become the leading consumer of Rheumatoid Arthritis Stem Cell Therapy by 2028?What manufacturing techniques are involved in the production of the Rheumatoid Arthritis Stem Cell Therapy?Which regions are the Rheumatoid Arthritis Stem Cell Therapy market players targeting to channelize their production portfolio?Get Full Access of the Report @https://www.factmr.com/report/1001/rheumatoid-arthritis-stem-cell-therapy-market
Pertinent aspects this study on the Rheumatoid Arthritis Stem Cell Therapy market tries to answer exhaustively are:
What is the forecast size (revenue/volumes) of the most lucrative regional market? What is the share of the dominant product/technology segment in the Rheumatoid Arthritis Stem Cell Therapy market? What regions are likely to witness sizable investments in research and development funding? What are Covid 19 implication on Rheumatoid Arthritis Stem Cell Therapy market and learn how businesses can respond, manage and mitigate the risks? Which countries will be the next destination for industry leaders in order to tap new revenue streams? Which new regulations might cause disruption in industry sentiments in near future? Which is the share of the dominant end user? Which region is expected to rise at the most dominant growth rate? Which technologies will have massive impact of new avenues in the Rheumatoid Arthritis Stem Cell Therapy market? Which key end-use industry trends are expected to shape the growth prospects of the Rheumatoid Arthritis Stem Cell Therapy market? What factors will promote new entrants in the Rheumatoid Arthritis Stem Cell Therapy market? What is the degree of fragmentation in the Rheumatoid Arthritis Stem Cell Therapy market, and will it increase in coming years?Why Choose Fact.MR?
Fact.MR follows a multi- disciplinary approach to extract information about various industries. Our analysts perform thorough primary and secondary research to gather data associated with the market. With modern industrial and digitalization tools, we provide avant-garde business ideas to our clients. We address clients living in across parts of the world with our 24/7 service availability.
Dana-Farber Patient Recovering Well After Cancer and the Coronavirus | Dana-Farber – Dana-Farber Cancer Institute
By daniellenierenberg
Pam Dobay is a warrior. In the last three years, the 67-year-old has dealt with a cancer diagnosis and stem cell transplant before recently contracting the coronavirus.
None of it was easy, but today, Dobay is recovering at home. She says she cannot begin to express the gratitude she feels towards everyone who has cared for her, including her Dana-Farber care team and her family.
When this is all over, I want to show everyone at Dana-Farber what they did, and thank them for everything, says Dobay.
In February 2018, Dobay was diagnosed with myelofibrosis, a blood disorder in which the bone marrow is unable to produce healthy red blood cells. Dobays primary care physician first worried something wasnt right after her test results from routine blood work came back abnormal. Myelofibrosis is a precursor condition for leukemia, meaning it puts those who are diagnosed at a much higher chance of developing the disease.
Dobay, who lives in Holbrook, MA, was placed under the care of Corey Cutler, MD, MPH, medical director of the Adult Stem Cell Transplantation Program at Dana-Farber/Brigham and Womens Cancer Center. Initially, she was given blood transfusions to help her body compensate for the bone marrows inability to produce red blood cells. This treatment is not designed to be a permanent fix, despite being highly effective for a short period of time: Eventually, Dobay would need a bone marrow transplant.
In September 2018, just six months after her diagnosis, Dobay underwent a reduced-intensity transplant (sometimes referred to as a mini-transplant). Mini-transplant patients receive lower doses of chemotherapy than are used in a full-intensity transplant, and in general, receive no radiation therapy. The reduced-intensity procedure was developed for older patients and others who often cant tolerate the harsh side effects of full-intensity treatments.
The procedure still proved to be difficult for Dobay, who ended up in the intensive care unit (ICU) due to complications. This was a possibility her care team had prepared for, and slowly, her condition improved. While she still has some symptoms of chronic graft-versus-host disease (GVHD), she and her family including Robert Dobay, her husband of 45 years hoped this would be her toughest test.
In March 2020, Dobay started experiencing fevers, chills, and difficulty breathing three symptoms of the coronavirus. Dobays family called Cutler, who instructed them to immediately bring her to the nearest emergency room. She was initially treated at her local hospital, but after she tested positive for the coronavirus, the family pushed for her to be transported to Brigham and Womens Hospital.
The team at Dana-Farber encourages anyone who is experiencing symptoms associated with COVID-19 to report them right away. Even if you have a confirmed case of the coronavirus, there are measures in place to ensure you can receive the care thats safe for you, your care team, and other patients and staff members.
Because Dobay was a former bone marrow transplant recipient and has GVHD, she is immunocompromisedand was at an increased risk for developing severe symptoms due to COVID-19. Upon being admitted to the ICU at Brigham and Womens, her condition worsened, and she needed to be placed on a ventilator.
Due to visitor restrictions, the Dobays were not allowed to visit her in the hospital, so her husband called her care team every day to check in.
Dr. Cutler contacted Francisco Marty, MD, an infectious disease specialist in the Adult Stem Cell Transplantation Center at Dana-Farber/Brigham and Womens Cancer Center, to discuss Dobays care. Marty was the principal investigator of trials testing the antiviral drug remdesivir, and its ability to treat patients with COVID-19 pneumonia.At the time, it was unclear if it would help bone marrow transplant recipients.
Dobay became the first stem cell transplant patient at Brigham and Womens to receive the drug. A week after starting treatment her lung function improved, and she was able to come off the ventilator. However, other medical complications led to her being placed back on the ventilator and to remain in the intensive care unit for a couple of more weeks. Over time, she was once again taken off the ventilator, and in early May she was finally able to go home.
We were very happy that the remdesivir trials were open to all patients, including many of our cancer and bone marrow transplant patients at Dana-Farber, says Marty. The initial reports from the clinical trials have shown patients who receive remdesivir recover faster from COVID-19 pneumonia. As additional analyses from the trials are performed, we will be able to see more closely how remdesivir helped the cancer patients at Dana-Farber and elsewhere.
We were all terrified, but she is just incredibly strong, Robert Dobay says. We are so thankful for her care team who helped her get through this.
Dobay says she has regained most of her strength since returning home, and in addition to her physical therapy, she is once again doing squats, weightlifting, and sit-ups. She is also back to going on walks with her daughter and her dog.
Pam has such a strong will, and her supportive husband was a remarkably large part of her recovery, says Tricia Severns, ANP-BC, OCN, a nurse practitioner at Dana-Farber and a member of Dobays care team. The entire family is incredibly strong and supportive.
I feel really lucky to still be here, adds Dobay. I could not have gotten through all of this without my family. Wed do anything for one another.
Excerpt from:
Dana-Farber Patient Recovering Well After Cancer and the Coronavirus | Dana-Farber - Dana-Farber Cancer Institute