Category Archives: Induced Pluripotent Stem Cells

Poultry Healthcare Products Market Share, Growth, Statistics, by Application, Production, Revenue & Forecast to 2025 – AlgosOnline

The ' Poultry Healthcare Products market' research report now available at MarketStudyReport.com delivers a thorough analysis of the industry trends influencing the global business scenario. In addition, the report offers definitive information pertaining to the commercialization aspects, revenue estimation, and market size of the industry. The report overtly defines the status of key players in the competitive landscape while including their portfolio and geographical expansion endeavors.

The latest research report on Poultry Healthcare Products market organizes updates pertaining to the growth catalysts, challenges, and opportunities that will define the industry growth over the forecast duration. In addition, it charts the course that business owners should take to address the changes brought in by the Covid-19 pandemic.

Request a sample Report of Poultry Healthcare Products Market at:https://www.marketstudyreport.com/request-a-sample/2545485?utm_source=algosonline.com&utm_medium=AG

Our analysts project that Poultry Healthcare Products market will register a CAGR of XX% over the forecast period 2020-2025.

Moreover, detailed analysis of each regional market as well as insights on the major players that occupy them are highlighted. Individual assessment of the industry segmentation and factors influencing their growth are extensively discussed for a stronger realization of top revenue prospects.

Market snapshot:

Regional outlook:

Product terrain outline:

Application spectrum overview:

Ask for Discount on Poultry Healthcare Products Market Report at:https://www.marketstudyreport.com/check-for-discount/2545485?utm_source=algosonline.com&utm_medium=AG

Competitive landscape summary:

Highlights of the Report:

For More Details On this Report: https://www.marketstudyreport.com/reports/global-poultry-healthcare-products-market-growth-status-and-outlook-2020-2025

Some of the Major Highlights of TOC covers:

Chapter 1: Methodology & Scope

Definition and forecast parameters

Methodology and forecast parameters

Data Sources

Chapter 2: Executive Summary

Business trends

Regional trends

Product trends

End-use trends

Chapter 3: Poultry Healthcare Products Industry Insights

Industry segmentation

Industry landscape

Vendor matrix

Technological and innovation landscape

Chapter 4: Poultry Healthcare Products Market, By Region

Chapter 5: Company Profile

Business Overview

Financial Data

Product Landscape

Strategic Outlook

SWOT Analysis

Related Reports:

2. Global Cutaneous Fibrosis Treatment Market Growth (Status and Outlook) 2021-2026 Cutaneous Fibrosis Treatment Market report covers the market landscape and its growth prospects over the coming years, the Report also brief deals with the product life cycle, comparing it to the relevant products from across industries that had already been commercialized details the potential for various applications, discussing about recent product innovations and gives an overview on potential regional market. Read More: https://www.marketstudyreport.com/reports/global-cutaneous-fibrosis-treatment-market-growth-status-and-outlook-2021-2026

Contact Us: Corporate Sales, Market Study Report LLC Phone: 1-302-273-0910 Toll Free: 1-866-764-2150 Email: [emailprotected]

See the original post here:
Poultry Healthcare Products Market Share, Growth, Statistics, by Application, Production, Revenue & Forecast to 2025 - AlgosOnline

Impact Of Covid 19 On Stem Cells Industry 2020 Market Challenges, Business Overview And Forecast Research Study 2026 Murphy’s Hockey Law – Murphy’s…

Overview for Stem Cells Market Helps in providing scope and definitions, Key Findings, Growth Drivers, and Various Dynamics.

Stem Cells Market Data and Acquisition Research Study with Trends and Opportunities 2019-2024 The study of Stem Cells market is a compilation of the market of Stem Cells broken down into its entirety on the basis of types, application, trends and opportunities, mergers and acquisitions, drivers and restraints, and a global outreach. The detailed study also offers a board interpretation of the Stem Cells industry from a variety of data points that are collected through reputable and verified sources. Furthermore, the study sheds a lights on a market interpretations on a global scale which is further distributed through distribution channels, generated incomes sources and a marginalized market space where most trade occurs.

Along with a generalized market study, the report also consists of the risks that are often neglected when it comes to the Stem Cells industry in a comprehensive manner. The study is also divided in an analytical space where the forecast is predicted through a primary and secondary research methodologies along with an in-house model.

Download PDF Sample of Stem Cells Market report @ https://hongchunresearch.com/request-a-sample/117744

Key players in the global Stem Cells market covered in Chapter 4: Bio-Rad Laboratories MEDIPOST Miltenyi Biotec Pluristem Therapeutics STEMCELL Technologies Becton, Dickinson and Company Holostem Terapie Avanzate Merck Group Cellular Dynamics International Lonza Group Thermo Fisher Scientific Anterogen Osiris Therapeutics Takara Bio Group Pharmicell

In Chapter 11 and 13.3, on the basis of types, the Stem Cells market from 2015 to 2026 is primarily split into: Adult Stem Cell Human Embryonic Stem Cell Induced Pluripotent Stem Cell Rat Neural Stem Cell Other

In Chapter 12 and 13.4, on the basis of applications, the Stem Cells market from 2015 to 2026 covers: Regenerative Medicine Drug Discovery and Development

Geographically, the detailed analysis of consumption, revenue, market share and growth rate, historic and forecast (2015-2026) of the following regions are covered in Chapter 5, 6, 7, 8, 9, 10, 13: North America (Covered in Chapter 6 and 13) United States Canada Mexico Europe (Covered in Chapter 7 and 13) Germany UK France Italy Spain Russia Others Asia-Pacific (Covered in Chapter 8 and 13) China Japan South Korea Australia India Southeast Asia Others Middle East and Africa (Covered in Chapter 9 and 13) Saudi Arabia UAE Egypt Nigeria South Africa Others South America (Covered in Chapter 10 and 13) Brazil Argentina Columbia Chile Others Regional scope can be customized

For a global outreach, the Stem Cells study also classifies the market into a global distribution where key market demographics are established based on the majority of the market share. The following markets that are often considered for establishing a global outreach are North America, Europe, Asia, and the Rest of the World. Depending on the study, the following markets are often interchanged, added, or excluded as certain markets only adhere to certain products and needs.

Here is a short glance at what the study actually encompasses: Study includes strategic developments, latest product launches, regional growth markers and mergers & acquisitions Revenue, cost price, capacity & utilizations, import/export rates and market share Forecast predictions are generated from analytical data sources and calculated through a series of in-house processes.

However, based on requirements, this report could be customized for specific regions and countries.

Brief about Stem Cells Market Report with [emailprotected]https://hongchunresearch.com/report/stem-cells-market-size-2020-117744

Some Point of Table of Content:

Chapter One: Report Overview

Chapter Two: Global Market Growth Trends

Chapter Three: Value Chain of Stem Cells Market

Chapter Four: Players Profiles

Chapter Five: Global Stem Cells Market Analysis by Regions

Chapter Six: North America Stem Cells Market Analysis by Countries

Chapter Seven: Europe Stem Cells Market Analysis by Countries

Chapter Eight: Asia-Pacific Stem Cells Market Analysis by Countries

Chapter Nine: Middle East and Africa Stem Cells Market Analysis by Countries

Chapter Ten: South America Stem Cells Market Analysis by Countries

Chapter Eleven: Global Stem Cells Market Segment by Types

Chapter Twelve: Global Stem Cells Market Segment by Applications 12.1 Global Stem Cells Sales, Revenue and Market Share by Applications (2015-2020) 12.1.1 Global Stem Cells Sales and Market Share by Applications (2015-2020) 12.1.2 Global Stem Cells Revenue and Market Share by Applications (2015-2020) 12.2 Regenerative Medicine Sales, Revenue and Growth Rate (2015-2020) 12.3 Drug Discovery and Development Sales, Revenue and Growth Rate (2015-2020)

Chapter Thirteen: Stem Cells Market Forecast by Regions (2020-2026) continued

Check [emailprotected] https://hongchunresearch.com/check-discount/117744

List of tables List of Tables and Figures Table Global Stem Cells Market Size Growth Rate by Type (2020-2026) Figure Global Stem Cells Market Share by Type in 2019 & 2026 Figure Adult Stem Cell Features Figure Human Embryonic Stem Cell Features Figure Induced Pluripotent Stem Cell Features Figure Rat Neural Stem Cell Features Figure Other Features Table Global Stem Cells Market Size Growth by Application (2020-2026) Figure Global Stem Cells Market Share by Application in 2019 & 2026 Figure Regenerative Medicine Description Figure Drug Discovery and Development Description Figure Global COVID-19 Status Overview Table Influence of COVID-19 Outbreak on Stem Cells Industry Development Table SWOT Analysis Figure Porters Five Forces Analysis Figure Global Stem Cells Market Size and Growth Rate 2015-2026 Table Industry News Table Industry Policies Figure Value Chain Status of Stem Cells Figure Production Process of Stem Cells Figure Manufacturing Cost Structure of Stem Cells Figure Major Company Analysis (by Business Distribution Base, by Product Type) Table Downstream Major Customer Analysis (by Region) Table Bio-Rad Laboratories Profile Table Bio-Rad Laboratories Production, Value, Price, Gross Margin 2015-2020 Table MEDIPOST Profile Table MEDIPOST Production, Value, Price, Gross Margin 2015-2020 Table Miltenyi Biotec Profile Table Miltenyi Biotec Production, Value, Price, Gross Margin 2015-2020 Table Pluristem Therapeutics Profile Table Pluristem Therapeutics Production, Value, Price, Gross Margin 2015-2020 Table STEMCELL Technologies Profile Table STEMCELL Technologies Production, Value, Price, Gross Margin 2015-2020 Table Becton, Dickinson and Company Profile Table Becton, Dickinson and Company Production, Value, Price, Gross Margin 2015-2020 Table Holostem Terapie Avanzate Profile Table Holostem Terapie Avanzate Production, Value, Price, Gross Margin 2015-2020 Table Merck Group Profile Table Merck Group Production, Value, Price, Gross Margin 2015-2020 Table Cellular Dynamics International Profile Table Cellular Dynamics International Production, Value, Price, Gross Margin 2015-2020 Table Lonza Group Profile Table Lonza Group Production, Value, Price, Gross Margin 2015-2020 Table Thermo Fisher Scientific Profile Table Thermo Fisher Scientific Production, Value, Price, Gross Margin 2015-2020 Table Anterogen Profile Table Anterogen Production, Value, Price, Gross Margin 2015-2020 Table Osiris Therapeutics Profile Table Osiris Therapeutics Production, Value, Price, Gross Margin 2015-2020 Table Takara Bio Group Profile Table Takara Bio Group Production, Value, Price, Gross Margin 2015-2020 Table Pharmicell Profile Table Pharmicell Production, Value, Price, Gross Margin 2015-2020 Figure Global Stem Cells Sales and Growth Rate (2015-2020) Figure Global Stem Cells Revenue ($) and Growth (2015-2020) Table Global Stem Cells Sales by Regions (2015-2020) Table Global Stem Cells Sales Market Share by Regions (2015-2020) Table Global Stem Cells Revenue ($) by Regions (2015-2020) Table Global Stem Cells Revenue Market Share by Regions (2015-2020) Table Global Stem Cells Revenue Market Share by Regions in 2015 Table Global Stem Cells Revenue Market Share by Regions in 2019 Figure North America Stem Cells Sales and Growth Rate (2015-2020) Figure Europe Stem Cells Sales and Growth Rate (2015-2020) Figure Asia-Pacific Stem Cells Sales and Growth Rate (2015-2020) Figure Middle East and Africa Stem Cells Sales and Growth Rate (2015-2020) Figure South America Stem Cells Sales and Growth Rate (2015-2020) Figure North America Stem Cells Revenue ($) and Growth (2015-2020) Table North America Stem Cells Sales by Countries (2015-2020) Table North America Stem Cells Sales Market Share by Countries (2015-2020) Figure North America Stem Cells Sales Market Share by Countries in 2015 Figure North America Stem Cells Sales Market Share by Countries in 2019 Table North America Stem Cells Revenue ($) by Countries (2015-2020) Table North America Stem Cells Revenue Market Share by Countries (2015-2020) Figure North America Stem Cells Revenue Market Share by Countries in 2015 Figure North America Stem Cells Revenue Market Share by Countries in 2019 Figure United States Stem Cells Sales and Growth Rate (2015-2020) Figure Canada Stem Cells Sales and Growth Rate (2015-2020) Figure Mexico Stem Cells Sales and Growth (2015-2020) Figure Europe Stem Cells Revenue ($) Growth (2015-2020) Table Europe Stem Cells Sales by Countries (2015-2020) Table Europe Stem Cells Sales Market Share by Countries (2015-2020) Figure Europe Stem Cells Sales Market Share by Countries in 2015 Figure Europe Stem Cells Sales Market Share by Countries in 2019 Table Europe Stem Cells Revenue ($) by Countries (2015-2020) Table Europe Stem Cells Revenue Market Share by Countries (2015-2020) Figure Europe Stem Cells Revenue Market Share by Countries in 2015 Figure Europe Stem Cells Revenue Market Share by Countries in 2019 Figure Germany Stem Cells Sales and Growth Rate (2015-2020) Figure UK Stem Cells Sales and Growth Rate (2015-2020) Figure France Stem Cells Sales and Growth Rate (2015-2020) Figure Italy Stem Cells Sales and Growth Rate (2015-2020) Figure Spain Stem Cells Sales and Growth Rate (2015-2020) Figure Russia Stem Cells Sales and Growth Rate (2015-2020) Figure Asia-Pacific Stem Cells Revenue ($) and Growth (2015-2020) Table Asia-Pacific Stem Cells Sales by Countries (2015-2020) Table Asia-Pacific Stem Cells Sales Market Share by Countries (2015-2020) Figure Asia-Pacific Stem Cells Sales Market Share by Countries in 2015 Figure Asia-Pacific Stem Cells Sales Market Share by Countries in 2019 Table Asia-Pacific Stem Cells Revenue ($) by Countries (2015-2020) Table Asia-Pacific Stem Cells Revenue Market Share by Countries (2015-2020) Figure Asia-Pacific Stem Cells Revenue Market Share by Countries in 2015 Figure Asia-Pacific Stem Cells Revenue Market Share by Countries in 2019 Figure China Stem Cells Sales and Growth Rate (2015-2020) Figure Japan Stem Cells Sales and Growth Rate (2015-2020) Figure South Korea Stem Cells Sales and Growth Rate (2015-2020) Figure Australia Stem Cells Sales and Growth Rate (2015-2020) Figure India Stem Cells Sales and Growth Rate (2015-2020) Figure Southeast Asia Stem Cells Sales and Growth Rate (2015-2020) Figure Middle East and Africa Stem Cells Revenue ($) and Growth (2015-2020) continued

About HongChun Research: HongChun Research main aim is to assist our clients in order to give a detailed perspective on the current market trends and build long-lasting connections with our clientele. Our studies are designed to provide solid quantitative facts combined with strategic industrial insights that are acquired from proprietary sources and an in-house model.

Contact Details: Jennifer Gray Manager Global Sales + 852 8170 0792 [emailprotected]

NOTE: Our report does take into account the impact of coronavirus pandemic and dedicates qualitative as well as quantitative sections of information within the report that emphasizes the impact of COVID-19.

As this pandemic is ongoing and leading to dynamic shifts in stocks and businesses worldwide, we take into account the current condition and forecast the market data taking into consideration the micro and macroeconomic factors that will be affected by the pandemic.

https://murphyshockeylaw.net/

Go here to read the rest:
Impact Of Covid 19 On Stem Cells Industry 2020 Market Challenges, Business Overview And Forecast Research Study 2026 Murphy's Hockey Law - Murphy's...

SREBP1 suppresses the differentiation and epithelial function of hiPSC-derived endothelial cells by inhibiting the microRNA199b-5p pathway – DocWire…

This article was originally published here

Stem Cell Res. 2021 Jan 13;51:102174. doi: 10.1016/j.scr.2021.102174. Online ahead of print.

ABSTRACT

Human induced pluripotent stem cell (hiPSC)-derived endothelial cell (hiPSC-EC) transplantation is a promising therapy for treating peripheral artery disease (PAD). However, the poor differentiation of hiPSCs limits their clinical application. Therefore, finding key factors that regulate cellular differentiation is crucial for improving the therapeutic efficacy of hiPSC-EC transplantation. Sterol regulatory element binding protein 1 (SREBP1) is a key regulator of lipid metabolism and stem cell differentiation. However, it remains unknown whether SREPBP1 modulates hiPSC differentiation. In this study, we showed that SREBP1 expression was negatively associated with hiPSC differentiation and EC function. The results show that SREBP1 binds to the promoter region of miR199b-5p and suppresses its transcription, resulting in the activation of Notch1 signaling. Blocking SREBP1 increased both hiPSC differentiation and EC angiogenesis. These findings demonstrate a novel role for SREBP1 in hiPSC differentiation and EC angiogenesis.

PMID:33485183 | DOI:10.1016/j.scr.2021.102174

Original post:
SREBP1 suppresses the differentiation and epithelial function of hiPSC-derived endothelial cells by inhibiting the microRNA199b-5p pathway - DocWire...

Induced Pluripotent Stem Cells Market Segmentation and Analysis by Latest Trends, Development and Growth by Trending Regions 2020 with key players…

The Induced Pluripotent Stem Cells Market grew in 2019, as compared to 2018, according to our report, Induced Pluripotent Stem Cells Market is likely to have subdued growth in 2020 due to weak demand on account of reduced industry spending post Covid-19 outbreak. Further, Induced Pluripotent Stem Cells Market will begin picking up momentum gradually from 2021 onwards and grow at a healthy CAGR between 2021-2025.

Deep analysis about Induced Pluripotent Stem Cells Market status (2016-2019), competition pattern, advantages and disadvantages of products, industry development trends (2019-2025), regional industrial layout characteristics and macroeconomic policies, industrial policy has also been included. From raw materials to downstream buyers of this industry have been analysed scientifically. This report will help you to establish comprehensive overview of the Induced Pluripotent Stem Cells Market

Get a Sample Copy of the Report at: https://i2iresearch.com/download-sample/?id=8913

The Induced Pluripotent Stem Cells Market is analysed based on product types, major applications and key players

Key product type: Hepatocytes Fibroblasts Keratinocytes Amniotic Cells Others

Key applications: Academic Research Drug Development And Discovery Toxicity Screening Regenerative Medicine

Key players or companies covered are: Fujifilm Holding Corporation Astellas Pharma Fate Therapeutics Bristol-Myers Squibb Company ViaCyte Celgene Corporation Aastrom Biosciences Acelity Holdings StemCells Japan Tissue Engineering Organogenesis

The report provides analysis & data at a regional level (North America, Europe, Asia Pacific, Middle East & Africa , Rest of the world) & Country level (13 key countries The U.S, Canada, Germany, France, UK, Italy, China, Japan, India, Middle East, Africa, South America)

Inquire or share your questions, if any: https://i2iresearch.com/need-customization/?id=8913

Key questions answered in the report: 1. What is the current size of the Induced Pluripotent Stem Cells Market, at a global, regional & country level? 2. How is the market segmented, who are the key end user segments? 3. What are the key drivers, challenges & trends that is likely to impact businesses in the Induced Pluripotent Stem Cells Market? 4. What is the likely market forecast & how will be Induced Pluripotent Stem Cells Market impacted? 5. What is the competitive landscape, who are the key players? 6. What are some of the recent M&A, PE / VC deals that have happened in the Induced Pluripotent Stem Cells Market?

The report also analysis the impact of COVID 19 based on a scenario-based modelling. This provides a clear view of how has COVID impacted the growth cycle & when is the likely recovery of the industry is expected to pre-covid levels.

Contact us: i2iResearch info to intelligence Locational Office: *India, *United States, *Germany Email: [emailprotected] Toll-free: +1-800-419-8865 | Phone: +91 98801 53667

https://murphyshockeylaw.net/

Read the original here:
Induced Pluripotent Stem Cells Market Segmentation and Analysis by Latest Trends, Development and Growth by Trending Regions 2020 with key players...

Induced Pluripotent Stem Cells (iPSCs) Market to Witness Massive Growth During 2021-2027 | Fujifilm Holding Corporation (CDI), Ncardia, Sumitomo…

LOS ANGELES, United States: QY Research has recently published a research report titled, Global Induced Pluripotent Stem Cells (iPSCs) Market Size, Status and Forecast 2021-2027. This report has been prepared by experienced and knowledgeable market analysts and researchers. It is a phenomenal compilation of important studies that explore the competitive landscape, segmentation, geographical expansion, and revenue, production, and consumption growth of the global Induced Pluripotent Stem Cells (iPSCs) market. Players can use the accurate market facts and figures and statistical studies provided in the report to understand the current and future growth of the global Induced Pluripotent Stem Cells (iPSCs) market.

The report includes CAGR, market shares, sales, gross margin, value, volume, and other vital market figures that give an exact picture of the growth of the global Induced Pluripotent Stem Cells (iPSCs) market.

Competitive Landscape

Competitor analysis is one of the best sections of the report that compares the progress of leading players based on crucial parameters, including market share, new developments, global reach, local competition, price, and production. From the nature of competition to future changes in the vendor landscape, the report provides in-depth analysis of the competition in the global Induced Pluripotent Stem Cells (iPSCs) market.

Key questions answered in the report:

TOC

1 Report Overview 1.1 Study Scope 1.2 Market Analysis by Type 1.2.1 Global Induced Pluripotent Stem Cells (iPSCs) Market Size Growth Rate by Type: 2016 VS 2021 VS 2027 1.2.2 Human iPSCs 1.2.3 Mouse iPSCs 1.3 Market by Application 1.3.1 Global Induced Pluripotent Stem Cells (iPSCs) Market Share by Application: 2016 VS 2021 VS 2027 1.3.2 Academic Research 1.3.3 Drug Development and Discovery 1.3.4 Toxicity Screening 1.3.5 Regenerative Medicine 1.3.6 Others 1.4 Study Objectives 1.5 Years Considered 2 Global Growth Trends 2.1 Global Induced Pluripotent Stem Cells (iPSCs) Market Perspective (2016-2027) 2.2 Induced Pluripotent Stem Cells (iPSCs) Growth Trends by Regions 2.2.1 Induced Pluripotent Stem Cells (iPSCs) Market Size by Regions: 2016 VS 2021 VS 2027 2.2.2 Induced Pluripotent Stem Cells (iPSCs) Historic Market Share by Regions (2016-2021) 2.2.3 Induced Pluripotent Stem Cells (iPSCs) Forecasted Market Size by Regions (2022-2027) 2.3 Induced Pluripotent Stem Cells (iPSCs) Industry Dynamic 2.3.1 Induced Pluripotent Stem Cells (iPSCs) Market Trends 2.3.2 Induced Pluripotent Stem Cells (iPSCs) Market Drivers 2.3.3 Induced Pluripotent Stem Cells (iPSCs) Market Challenges 2.3.4 Induced Pluripotent Stem Cells (iPSCs) Market Restraints 3 Competition Landscape by Key Players 3.1 Global Top Induced Pluripotent Stem Cells (iPSCs) Players by Revenue 3.1.1 Global Top Induced Pluripotent Stem Cells (iPSCs) Players by Revenue (2016-2021) 3.1.2 Global Induced Pluripotent Stem Cells (iPSCs) Revenue Market Share by Players (2016-2021) 3.2 Global Induced Pluripotent Stem Cells (iPSCs) Market Share by Company Type (Tier 1, Tier 2 and Tier 3) 3.3 Players Covered: Ranking by Induced Pluripotent Stem Cells (iPSCs) Revenue 3.4 Global Induced Pluripotent Stem Cells (iPSCs) Market Concentration Ratio 3.4.1 Global Induced Pluripotent Stem Cells (iPSCs) Market Concentration Ratio (CR5 and HHI) 3.4.2 Global Top 10 and Top 5 Companies by Induced Pluripotent Stem Cells (iPSCs) Revenue in 2020 3.5 Induced Pluripotent Stem Cells (iPSCs) Key Players Head office and Area Served 3.6 Key Players Induced Pluripotent Stem Cells (iPSCs) Product Solution and Service 3.7 Date of Enter into Induced Pluripotent Stem Cells (iPSCs) Market 3.8 Mergers & Acquisitions, Expansion Plans 4 Induced Pluripotent Stem Cells (iPSCs) Breakdown Data by Type 4.1 Global Induced Pluripotent Stem Cells (iPSCs) Historic Market Size by Type (2016-2021) 4.2 Global Induced Pluripotent Stem Cells (iPSCs) Forecasted Market Size by Type (2022-2027) 5 Induced Pluripotent Stem Cells (iPSCs) Breakdown Data by Application 5.1 Global Induced Pluripotent Stem Cells (iPSCs) Historic Market Size by Application (2016-2021) 5.2 Global Induced Pluripotent Stem Cells (iPSCs) Forecasted Market Size by Application (2022-2027) 6 North America 6.1 North America Induced Pluripotent Stem Cells (iPSCs) Market Size (2016-2027) 6.2 North America Induced Pluripotent Stem Cells (iPSCs) Market Size by Type 6.2.1 North America Induced Pluripotent Stem Cells (iPSCs) Market Size by Type (2016-2021) 6.2.2 North America Induced Pluripotent Stem Cells (iPSCs) Market Size by Type (2022-2027) 6.2.3 North America Induced Pluripotent Stem Cells (iPSCs) Market Size by Type (2016-2027) 6.3 North America Induced Pluripotent Stem Cells (iPSCs) Market Size by Application 6.3.1 North America Induced Pluripotent Stem Cells (iPSCs) Market Size by Application (2016-2021) 6.3.2 North America Induced Pluripotent Stem Cells (iPSCs) Market Size by Application (2022-2027) 6.3.3 North America Induced Pluripotent Stem Cells (iPSCs) Market Size by Application (2016-2027) 6.4 North America Induced Pluripotent Stem Cells (iPSCs) Market Size by Country 6.4.1 North America Induced Pluripotent Stem Cells (iPSCs) Market Size by Country (2016-2021) 6.4.2 North America Induced Pluripotent Stem Cells (iPSCs) Market Size by Country (2022-2027) 6.4.3 United States 6.4.3 Canada 7 Europe 7.1 Europe Induced Pluripotent Stem Cells (iPSCs) Market Size (2016-2027) 7.2 Europe Induced Pluripotent Stem Cells (iPSCs) Market Size by Type 7.2.1 Europe Induced Pluripotent Stem Cells (iPSCs) Market Size by Type (2016-2021) 7.2.2 Europe Induced Pluripotent Stem Cells (iPSCs) Market Size by Type (2022-2027) 7.2.3 Europe Induced Pluripotent Stem Cells (iPSCs) Market Size by Type (2016-2027) 7.3 Europe Induced Pluripotent Stem Cells (iPSCs) Market Size by Application 7.3.1 Europe Induced Pluripotent Stem Cells (iPSCs) Market Size by Application (2016-2021) 7.3.2 Europe Induced Pluripotent Stem Cells (iPSCs) Market Size by Application (2022-2027) 7.3.3 Europe Induced Pluripotent Stem Cells (iPSCs) Market Size by Application (2016-2027) 7.4 Europe Induced Pluripotent Stem Cells (iPSCs) Market Size by Country 7.4.1 Europe Induced Pluripotent Stem Cells (iPSCs) Market Size by Country (2016-2021) 7.4.2 Europe Induced Pluripotent Stem Cells (iPSCs) Market Size by Country (2022-2027) 7.4.3 Germany 7.4.4 France 7.4.5 U.K. 7.4.6 Italy 7.4.7 Russia 7.4.8 Nordic 8 Asia-Pacific 8.1 Asia-Pacific Induced Pluripotent Stem Cells (iPSCs) Market Size (2016-2027) 8.2 Asia-Pacific Induced Pluripotent Stem Cells (iPSCs) Market Size by Type 8.2.1 Asia-Pacific Induced Pluripotent Stem Cells (iPSCs) Market Size by Type (2016-2021) 8.2.2 Asia-Pacific Induced Pluripotent Stem Cells (iPSCs) Market Size by Type (2022-2027) 8.2.3 Asia-Pacific Induced Pluripotent Stem Cells (iPSCs) Market Size by Type (2016-2027) 8.3 Asia-Pacific Induced Pluripotent Stem Cells (iPSCs) Market Size by Application 8.3.1 Asia-Pacific Induced Pluripotent Stem Cells (iPSCs) Market Size by Application (2016-2021) 8.3.2 Asia-Pacific Induced Pluripotent Stem Cells (iPSCs) Market Size by Application (2022-2027) 8.3.3 Asia-Pacific Induced Pluripotent Stem Cells (iPSCs) Market Size by Application (2016-2027) 8.4 Asia-Pacific Induced Pluripotent Stem Cells (iPSCs) Market Size by Region 8.4.1 Asia-Pacific Induced Pluripotent Stem Cells (iPSCs) Market Size by Region (2016-2021) 8.4.2 Asia-Pacific Induced Pluripotent Stem Cells (iPSCs) Market Size by Region (2022-2027) 8.4.3 China 8.4.4 Japan 8.4.5 South Korea 8.4.6 Southeast Asia 8.4.7 India 8.4.8 Australia 9 Latin America 9.1 Latin America Induced Pluripotent Stem Cells (iPSCs) Market Size (2016-2027) 9.2 Latin America Induced Pluripotent Stem Cells (iPSCs) Market Size by Type 9.2.1 Latin America Induced Pluripotent Stem Cells (iPSCs) Market Size by Type (2016-2021) 9.2.2 Latin America Induced Pluripotent Stem Cells (iPSCs) Market Size by Type (2022-2027) 9.2.3 Latin America Induced Pluripotent Stem Cells (iPSCs) Market Size by Type (2016-2027) 9.3 Latin America Induced Pluripotent Stem Cells (iPSCs) Market Size by Application 9.3.1 Latin America Induced Pluripotent Stem Cells (iPSCs) Market Size by Application (2016-2021) 9.3.2 Latin America Induced Pluripotent Stem Cells (iPSCs) Market Size by Application (2022-2027) 9.3.3 Latin America Induced Pluripotent Stem Cells (iPSCs) Market Size by Application (2016-2027) 9.4 Latin America Induced Pluripotent Stem Cells (iPSCs) Market Size by Country 9.4.1 Latin America Induced Pluripotent Stem Cells (iPSCs) Market Size by Country (2016-2021) 9.4.2 Latin America Induced Pluripotent Stem Cells (iPSCs) Market Size by Country (2022-2027) 9.4.3 Mexico 9.4.4 Brazil 10 Middle East & Africa 10.1 Middle East & Africa Induced Pluripotent Stem Cells (iPSCs) Market Size (2016-2027) 10.2 Middle East & Africa Induced Pluripotent Stem Cells (iPSCs) Market Size by Type 10.2.1 Middle East & Africa Induced Pluripotent Stem Cells (iPSCs) Market Size by Type (2016-2021) 10.2.2 Middle East & Africa Induced Pluripotent Stem Cells (iPSCs) Market Size by Type (2022-2027) 10.2.3 Middle East & Africa Induced Pluripotent Stem Cells (iPSCs) Market Size by Type (2016-2027) 10.3 Middle East & Africa Induced Pluripotent Stem Cells (iPSCs) Market Size by Application 10.3.1 Middle East & Africa Induced Pluripotent Stem Cells (iPSCs) Market Size by Application (2016-2021) 10.3.2 Middle East & Africa Induced Pluripotent Stem Cells (iPSCs) Market Size by Application (2022-2027) 10.3.3 Middle East & Africa Induced Pluripotent Stem Cells (iPSCs) Market Size by Application (2016-2027) 10.4 Middle East & Africa Induced Pluripotent Stem Cells (iPSCs) Market Size by Country 10.4.1 Middle East & Africa Induced Pluripotent Stem Cells (iPSCs) Market Size by Country (2016-2021) 10.4.2 Middle East & Africa Induced Pluripotent Stem Cells (iPSCs) Market Size by Country (2022-2027) 10.4.3 Turkey 10.4.4 Saudi Arabia 10.4.5 UAE 11 Key Players Profiles 11.1 Fujifilm Holding Corporation (CDI) 11.1.1 Fujifilm Holding Corporation (CDI) Company Details 11.1.2 Fujifilm Holding Corporation (CDI) Business Overview 11.1.3 Fujifilm Holding Corporation (CDI) Induced Pluripotent Stem Cells (iPSCs) Introduction 11.1.4 Fujifilm Holding Corporation (CDI) Revenue in Induced Pluripotent Stem Cells (iPSCs) Business (2016-2021) 11.1.5 Fujifilm Holding Corporation (CDI) Recent Development 11.2 Ncardia 11.2.1 Ncardia Company Details 11.2.2 Ncardia Business Overview 11.2.3 Ncardia Induced Pluripotent Stem Cells (iPSCs) Introduction 11.2.4 Ncardia Revenue in Induced Pluripotent Stem Cells (iPSCs) Business (2016-2021) 11.2.5 Ncardia Recent Development 11.3 Sumitomo Dainippon Pharma 11.3.1 Sumitomo Dainippon Pharma Company Details 11.3.2 Sumitomo Dainippon Pharma Business Overview 11.3.3 Sumitomo Dainippon Pharma Induced Pluripotent Stem Cells (iPSCs) Introduction 11.3.4 Sumitomo Dainippon Pharma Revenue in Induced Pluripotent Stem Cells (iPSCs) Business (2016-2021) 11.3.5 Sumitomo Dainippon Pharma Recent Development 11.4 Astellas Pharma Inc 11.4.1 Astellas Pharma Inc Company Details 11.4.2 Astellas Pharma Inc Business Overview 11.4.3 Astellas Pharma Inc Induced Pluripotent Stem Cells (iPSCs) Introduction 11.4.4 Astellas Pharma Inc Revenue in Induced Pluripotent Stem Cells (iPSCs) Business (2016-2021) 11.4.5 Astellas Pharma Inc Recent Development 11.5 Fate Therapeutics, Inc 11.5.1 Fate Therapeutics, Inc Company Details 11.5.2 Fate Therapeutics, Inc Business Overview 11.5.3 Fate Therapeutics, Inc Induced Pluripotent Stem Cells (iPSCs) Introduction 11.5.4 Fate Therapeutics, Inc Revenue in Induced Pluripotent Stem Cells (iPSCs) Business (2016-2021) 11.5.5 Fate Therapeutics, Inc Recent Development 11.6 Pluricell Biotech 11.6.1 Pluricell Biotech Company Details 11.6.2 Pluricell Biotech Business Overview 11.6.3 Pluricell Biotech Induced Pluripotent Stem Cells (iPSCs) Introduction 11.6.4 Pluricell Biotech Revenue in Induced Pluripotent Stem Cells (iPSCs) Business (2016-2021) 11.6.5 Pluricell Biotech Recent Development 11.7 Cell Inspire Biotechnology 11.7.1 Cell Inspire Biotechnology Company Details 11.7.2 Cell Inspire Biotechnology Business Overview 11.7.3 Cell Inspire Biotechnology Induced Pluripotent Stem Cells (iPSCs) Introduction 11.7.4 Cell Inspire Biotechnology Revenue in Induced Pluripotent Stem Cells (iPSCs) Business (2016-2021) 11.7.5 Cell Inspire Biotechnology Recent Development 11.8 ReproCELL 11.8.1 ReproCELL Company Details 11.8.2 ReproCELL Business Overview 11.8.3 ReproCELL Induced Pluripotent Stem Cells (iPSCs) Introduction 11.8.4 ReproCELL Revenue in Induced Pluripotent Stem Cells (iPSCs) Business (2016-2021) 11.8.5 ReproCELL Recent Development 12 Analysts Viewpoints/Conclusions 13 Appendix 13.1 Research Methodology 13.1.1 Methodology/Research Approach 13.1.2 Data Source 13.2 Disclaimer 13.3 Author Details

About Us:

QYResearch always pursuits high product quality with the belief that quality is the soul of business. Through years of effort and supports from huge number of customer supports, QYResearch consulting group has accumulated creative design methods on many high-quality markets investigation and research team with rich experience. Today, QYResearch has become the brand of quality assurance in consulting industry.

Go here to see the original:
Induced Pluripotent Stem Cells (iPSCs) Market to Witness Massive Growth During 2021-2027 | Fujifilm Holding Corporation (CDI), Ncardia, Sumitomo...

Hands across the water as bio bonds stand test of time | Business Weekly – Business Weekly

Boris Johnsons doomed reliance on Donald Trump to deliver a massive US trade deal for the UK is in stark contrast to the biotechnology bonds that continue to endure with massive commercial payback across the Atlantic.

Cambridge-US life science collaborations and investments have hit record levels in the last year and seemed destined to gather further pace through 2021 and beyond.

The Cluster now has GW Pharma and Bicycle Therapeutics under their own steam, Horizon Discovery (courtesy its acquisition by PerkinElmer), F-star (via its merger with Spring Bank) and Kymab (through its acquisition by Sanofi) quoted on Nasdaq, the US technology exchange. So lets delve deeper into the special relationship between UK and US biotechs that is not sullied by political fluctuations.

We reported in February 2020 that Bicycle Therapeutics clinched a $30 million upfront payment from Roche Group business Genentech as part of a strategic technology collaboration that could haul in up to $1.7 billion.

Bicycle will also be eligible to receive tiered royalties on Bicycle-based medicines commercialised by Genentech. Bicycle will be exploring its technology on a wider range of immuno-oncology targets, combining the expertise of both companies.

Bicycle will contribute its proprietary discovery platform, which allows rapid screening of novel targets to identify Bicycles and the ability to readily conjugate these together to create novel molecules that may overcome the potential limitations of other modalities. Genentech brings to the table its knowledge of immuno-oncology drug discovery and emerging target biology, as well as its development and commercialisation expertise.

In April 2020 and in another world first for a Cambridge life science business CellCentric revealed that it had developed CCS1477 the first p300/CBP inhibitor of its kind for use in the treatment of multiple cancer types.

Expanding on its UK-based clinical activities, the company disclosed that it was set to open US clinical sites for patient recruitment and had signed a milestone agreement with the Prostate Cancer Clinical Trial Consortium, LLC headquartered in New York to help select and manage US sites for the ongoing evaluation of CCS1477.

Also last April, Mogrify in the UK and Sangamo in the US agreed a collaboration and exclusive licence agreement for Mogrifys iPSC and ESC-derived regulatory T cells. It is a fistful of dollars play for startup business Mogrify which aims to transform the development of cell therapies by the systematic discovery of novel cell conversions; Nasdaq-quoted Sangamo Therapeutics is a genomic medicine company.

The deal allows Sangamo to develop allogeneic cell therapies from Mogrifys proprietary induced pluripotent stem cells and embryonic stem cells and Sangamos zinc finger protein gene-engineered chimeric antigen receptor regulatory T cell (CAR-Treg) technology.

In May, global food nutrition gamechanger Cargill headquartered in Minneapolis and Cambridge-based Eagle Genomics agreed a multi-year platform engagement to enable the digital transformation of microbiome and life sciences R & D across Cargills international territories. The deployment of the Eagle Genomics e[datascientist] platform will initially enable Cargills Health Technologies business to organise and synthesise additional insights from microbiome data amassed by the company over the past decade.

We announced in June that a young but big hitting US life science player was growing a UK team from a new base at Granta Park in Cambridge. Alloy Therapeutics, formed in Massachusetts in 2017, is dedicated to empowering global scientists with foundational drug discovery platforms and services. It is hiring fresh talent at its new UK home at Grantas McLintock Building.

The executive team is power-packed with business builders and money magnets. Led by founder, CEO and chairman Errik Anderson, the company is pioneering technology in a key growth area of the life sciences. Its specialist Discovery Services teams in Boston, MA and Cambridge, UK do immunisations every day while working to refine and improve the companys groundbreaking Alloy-Gx platform.

The sister Alloy Discovery Services business enables Alloy to ensure that all its partners are successful in their antibody discovery projects.

Also in June, Sosei Heptares set up a potential $409m exclusive discovery collaboration deal with New York-quoted AbbVie and there could be more cash cream to pour on top.

Sosei Heptares is eligible to receive up to $32m in upfront and near-term milestone payments as well as potential option, development and commercial milestones of up to $377m plus tiered royalties on global commercial sales.

Inivata, a Cambridge UK leader in liquid biopsy, reported in July that NeoGenomics, Inc its strategic commercialisation partner in the United States for the InVisionFirst-Lung liquid biopsy had commercially launched the test in the US.

InVisionFirst-Lung is a ctDNA next-generation sequencing liquid biopsy assay testing 37 genes relevant to the care of advanced non-small cell lung cancer. Nasdaq-quoted NeoGenomics, based in Florida, is a leading US-based cancer diagnostics and services company anchored in Florida.

July was also a seminal month for Acacia Pharma as the FDA approved its BYFAVO (remimazolam) for injection for the induction and maintenance of procedural sedation in adults undergoing surgical procedures lasting 30 minutes or less.

Acacia in-licensed the commercial rights to BYFAVO for the US from Cosmo Pharmaceuticals NV in January.

Avacta Group revealed in August that it could earn anything from $1/2bn to $1bn from an extended partnership with LG Chem Life Science, the Massachusetts-based subsidiary of the South Korean LG Group. The windfall stems from additional drug development programmes utilising Avactas Affimer XT technology.

Also in August, a US company personalising immune therapies for solid tumour cancers raised further capital to complete the Phase 1 buildout of a UK manufacturing facility in Sawston, Cambridge.

Northwest Biotherapeutics in Maryland raised $5 million growth capital and topped this up by securing a special purpose competitive loan of $1.77m from the Department for Business, Energy & Industrial Strategy. Northwest is developing DCVax personalised immune therapies for solid tumour cancers and is pursuing an intensive program of manufacturing preparations and planning as it approaches top line data from its Phase III trial.

In September, PhoreMost, the Cambridge biopharma company dedicated to drugging undruggable disease targets, and XtalPi Inc., a leading algorithm-driven AI-based pharmaceutical technology company, entered into a drug discovery collaboration agreement.

PhoreMost and Massachusetts-based XtalPi intend to rapidly identify and develop compounds to advance a drug discovery program against targets that epigenetically regulate tumour progression, and have been previously classified as undruggable.

A collaboration between Cambridge business Evonetix and Massachusetts company Analog Devices Inc was unveiled in September to fast-track the development and scale-up of the UK innovators desktop DNA writer. The technology will help facilitate the rapidly growing multibillion-dollar synthetic biology industry, Evonetix says.

Sosei Heptares earned more megabucks in September from its ongoing collaboration with $52 billion turnover New York-rooted pharma giant Pfizer. Pfizer reported that the first subject in a clinical trial had been dosed with a new drug candidate nominated from the multi-target drug discovery collaboration between the companies.

The milestone triggered a payment of $5 million to Sosei Heptares. The candidate was nominated for advancement by Pfizer in June 2019 generating a $3m milestone payment at that time.

Abzena pumped $60m into a new San Diego manufacturing hub in October a move that created 125 additional jobs with more to come.

Also last autumn, Cambridge biopharma business Arecor announced that it was partnering with global big-hitter Hikma Pharmaceuticals PLC to co-create a ready-to-administer injectable medicine in the US.

The breakthrough technology is being delivered through the quoted UK giants affiliate Hikma Pharmaceuticals USA Inc.

In December, Abcam opened a new 16,000 sq ft purpose-fitted facility in Fremont, CA, which will serve as a major new site for its specialist cell engineering team.

Also before Christmas, Owlstone Medical revealed that it was establishing a permanent office on the North Carolina Research Triangle Park and was hiring on both sides of the Atlantic.

Read more from the original source:
Hands across the water as bio bonds stand test of time | Business Weekly - Business Weekly

Exercise mimetics and JAK inhibition attenuate IFN-induced wasting in engineered human skeletal muscle – Science Advances

INTRODUCTION

Skeletal muscle comprises 45% of the total human body mass, and its contractile function is fundamental for the maintenance of life. Healthy skeletal muscle has a robust capacity to regenerate in response to minor injuries via activation, proliferation, and differentiation of muscle stem cells, a process that is greatly aided with the local and systemic inflammatory response (1). Interferon- (IFN-), in particular, is an important proinflammatory cytokine that regulates myogenic process during muscle regeneration, and its production is well balanced among natural killer (NK) cells, CD4+ and CD8+ T cells, and regulatory T cells (2). While inflammation following muscle injury promotes muscle regeneration, unregulated inflammatory reactions in many diseases, including chronic obstructive pulmonary disease, rheumatoid arthritis, dermatomyositis, cachexia, or sarcopenia, are associated with muscle loss and weakness (36). For example, sarcopenia is an age-associated syndrome characterized by progressive and generalized loss of skeletal muscle function and is usually associated with elevated expression of cytokines, including IFN- (6). In addition, elevated IFN- levels are routinely observed after influenza virus infection (7) and are associated with a cytokine storm resulting in tissue and organ damage in COVID-19 (8).

Besides numerous studies in rodents that show myopathic effects of inflammation in general and IFN- in particular (914), clinical studies have suggested that elevated IFN- in chronic inflammation and autoimmune diseases is one of major contributors to human skeletal muscle wasting and dysfunction (5, 6, 15). In a handful of in vitro studies in human muscle cells, IFN- has been shown to increase expression of human leukocyte antigenDR isotype (HLA-DR) class II antigens (16), proteases cathepsin B and L (17), and cytokines CXCL9, CXCL10, interleukin-6 (IL-6), and transforming growth factor (TGF-) (18, 19), as well as to inhibit myoblast growth and fusion (20). On the other hand, specific functional consequences and underlying mechanisms of IFN- elevation in human skeletal muscle have not been previously studied in vitro due to shortcomings of two-dimensional (2D) muscle cell culture (21). Recently, we have reported the first generation of 3D tissue-engineered models of functional human skeletal muscle (myobundles) made using primary myoblasts or induced pluripotent stem cellderived myogenic progenitors (2224). In optimized culture conditions, 3D human myobundles show superior myotube differentiation than 2D cultures and recapitulate the hallmark functional properties of native skeletal muscle including electrically or chemically induced twitch and tetanic contractions, a positive force-frequency relationship, robust calcium transients, and physiological responses to a diverse set of chemicals and drugs.

In more recent studies, we have shown that myobundles exhibit physiological response to exercise-mimetic electrical stimulation (E-stim) as evidenced from increased contractile strength, bulk muscle size, myotube diameter and length, sarcomeric protein expression, and glycolytic and fatty acid metabolism (24). In humans, E-stim and physical exercise can increase muscle mass and strength (25) and provide therapeutic benefits in the setting of chronic inflammation and aging (2628). The anti-inflammatory effects of muscle exercise are usually attributed to paracrine action of working myofibers on nonmuscle cells including adipocytes, macrophages, T cells, and NK cells (29). On the other hand, whether exercised myofibers can exert cell-autonomous anti-inflammatory effects on muscle structure and function has not been previously studied.

Here, we used the human myobundle system devoid of complex organ-organ interactions present in vivo to first time explore direct effects of IFN- on human skeletal muscle structure, function, and cytokine secretion and to unveil potential muscle-autonomous mechanisms underlying anti-inflammatory roles of exercise. We found that IFN- treatment induced muscle atrophy and reduced contractile function in human myobundles via up-regulation of the JAK/STAT (Janus kinase/signal transducer and activator of transcription) pathway (30), an effect that was directly countered by exercise-mimetic muscle stimulation. We further showed that the block of JAK/STAT up-regulation by IFN- with small-molecule JAK inhibitors tofacitinib (31) and baricitinib (32), clinically used for rheumatoid arthritis and in trials for COVID-19 (33), fully prevented muscle wasting and weakness induced by IFN-. Our findings establish human myobundles as a novel in vitro platform for studies of inflammatory muscle disease.

On the basis of previous studies in mice (9), we set to explore whether chronic 7-day application of a proinflammatory cytokine IFN- (20 ng/ml) to human myobundles cultured in serum-free media would lead to induction of muscle atrophy and weakness. Human myobundles made using cells derived from three independent donors were differentiated for 1 week, then exposed to IFN- for 1 week, and assessed for changes in cytokine secretion, structural, biochemical, and functional properties (Fig. 1A). To assess whether exercise-mimetic activity of myobundles can reduce expected myopathic effects of IFN-, we simultaneously applied chronic intermittent E-stim regime shown to increase myobundle mass and strength in our previous study (Fig. 1B) (24). In isometric force tests, we found that compared with the untreated control, IFN- treatment reduced myobundle twitch and tetanic force amplitude by 66 and 68%, respectively (Fig. 1, C, D, and G), while applying E-stim to untreated myobundles doubled their strength (Fig. 1, C, E, and G). E-stim cotreatment with IFN- significantly increased myobundle contractile force compared with IFN-only application (Fig. 1, D, F, and G), with twitch and tetanus amplitudes approximating those of untreated myobundles exposed to E-stim (Fig. 1, C, F, and G). These functional results were highly reproducible for myobundles from all three donors (fig. S1, A to C), as was the lack of any effects of IFN- or E-stim on passive tension measured at different stretch levels (fig. S1, D to F). Furthermore, measurements of force kinetics revealed that IFN- slowed both contraction (Fig. 1H) and relaxation (Fig. 1I) of myobundles and that E-stim rescued the IFN-induced force relaxation deficit as evidenced from the return of twitch recovery time (RT1/2) to control value (Fig. 1I). The exact numerical results of functional measurements are presented in table S1. Together, these studies showed that IFN- exerts adverse, inflammatory effects on contractile function of human myobundles and that exercise-mimetic activity can prevent IFN-induced muscle weakness.

(A) Schematic overview of myobundle culture, treatment, and characterization. Human primary myoblasts were expanded in 2D culture and mixed with hydrogel to form 3D myobundles, which were cultured in growth media (GM) for 4 days, then in differentiation media (DM) for 7 days, after which E-stim and/or IFN- (20 ng/ml) was applied for an additional 7 days. (B) E-stim protocol consisted of alternating 1-hour stimulation (S) at 10 Hz and 7-hour rest (R). (C to F) Representative twitch (Twi, 1 Hz) and tetanus (Tet, 20 Hz) force traces from myobundles (C) without IFN- or E-stim (IFN-, E-stim), (D) IFN-+, E-stim, (E) IFN-, E-stim+, and (F) IFN-+, E-stim+. (G) Twitch and tetanic force amplitudes averaged over three independent donors and shown relative to the IFN-, E-stim group (n = 5 to 10 myobundles per donor, 20 to 23 myobundles per group). (H and I) Time-to-peak tension (H; TPT) and half-relaxation time (I; 1/2RT) derived from contractile force recordings in myobundles (n = 47 to 75 data points from 20 to 23 myobundles from three donors per group). *P < 0.05 versus IFN-, #P < 0.05 versus IFN-+. NS, nonsignificant. Data are presented as means SEM.

We next investigated mechanisms underlying IFN-induced myobundle weakness and functional benefit of E-stim, including possible changes in muscle structure, cytokine secretion, and calcium handling. From cross-sectional stainings (Fig. 2, A to C, and fig. S2, A to C), we found that both IFN- and E-stim similarly increased myobundle cross-sectional area (CSA) by ~40%, while E-stim of IFN-treated myobundles did not induce additional CSA increase (Fig. 2D). Similar to our previous study (24), the number of nuclei in myobundle cross section was significantly increased by E-stim in the absence of IFN- (Fig. 2E), while application of IFN-, with or without E-stim, resulted in unchanged nuclear numbers. Total F-actin+ area, a measure of myobundle muscle mass, was not altered by IFN-, indicating that IFN-induced force deficit was not a result of total muscle mass loss (Fig. 2F). On the other hand, E-stim induced muscle mass increase, albeit more in untreated (F-actin+ area, 0.56 mm2) than in IFN-treated (0.43 mm2) myobundles (Fig. 2F). We then assessed possible alterations in the myotube size by staining myobundle cross sections for dystrophin, a myotube membrane protein (Fig. 2C and fig. S2C). The IFN- treatment significantly reduced myotube diameter (11.3-m untreated versus 8.8-m IFN- treated) and area (fig. S2D), thus inducing myotube atrophy, whereas E-stim induced myotube hypertrophy by increasing myotube diameter and area in untreated and, to less extent, IFN-treated myobundles (Fig. 2G and fig. S2D). We further examined density of myotubes [labeled by F-actin and sarcomeric -actinin (SAA)] in myobundle cross sections and found that IFN- decreased myotube density, which remained at control levels in the presence of E-stim (fig. S2, E and F). Numerical results are presented in table S1. Together, these analyses suggest that IFN-induced myotube size and density decrease were prevented in exercise-mimetic E-stim conditions. We also assessed specific force of myobundles (contractile force normalized by muscle CSA), and similar to effects on myotube size and density, we found that E-stim prevented IFN-induced force decrease (Fig. 2H).

E-Stim prevents IFN-induced structural deterioration of myobundles. (A to C) Representative myobundle cross sections immunostained for (A) F-actin (green; scale bars, 500 m), (B) sarcomeric -actinin (SAA; red; scale bars, 500 m), and (C) dystrophin (Dys; red; scale bars, 25 m). (D to G) Quantification of (D) myobundle cross-sectional area (CSA), (E) number of nuclei per cross section, (F) F-actin+ area, and (G) myotube diameter [n = 19 to 27 images from three donors per group, 3 to 5 myobundles for each donor for (D) to (F), n > 900 myotubes from three donors per group for (G)]. (H) Force amplitude normalized per myobundle CSA (specific force, n = 20 to 23 myobundles from three donors per group). *P < 0.05 versus IFN-, #P < 0.05 versus IFN-+, **P < 0.05. NS, nonsignificant. Data are presented as means SEM.

We also cryosectioned myobundles longitudinally and immunostained them for F-actin and SAA to assess potential changes in myotube and sarcomere organization across the entire myobundle depth (Fig. 3, A and B, and figs. S3 and S4). Adverse effects of IFN- treatment were most apparent in the interior of myobundles as evident from the decrease in projected myotube length (L; Fig. 3C and fig. S3, A and B), alignment (movie S1), and sarcomeric organization with significantly reduced percentage of cross-striated myotubes (from 45% in IFN- to 15% in IFN-+ group; Fig. 3D and fig. S3, C to F). As in our previous study (24), E-stim significantly increased myotube length (Fig. 3C) and percentage of cross-striated myofibers in untreated and, to a less extent, in IFN-treated myobundles (Fig. 3D), preventing the deteriorating effects of IFN- on myobundle structure. We then assessed changes in expression of contractile proteins (Fig. 3E) in myobundles and found that dystrophin and SAA expression remained unaltered with IFN- and E-stim treatments (Fig. 3, E to G), while changes in total myosin light chain (MYL) and myosin heavy chain (MYH) expression followed the same trend as the changes in measured specific force (Fig. 3, E, H, and I). Specifically, MYL and MYH protein expressions were significantly reduced by IFN- treatment and were increased by E-stim in both untreated and, to a less extent, IFN-treated myobundles. Regulation of MYL expression by skeletal muscle MYL kinase is associated with muscle differentiation (34) and the kinetics and amplitude of contractile force generation (35), while increase in MYH is associated with muscle differentiation, hypertrophy, and increased strength (24). Together, these studies showed that functional deficit due to IFN- treatment and benefit from E-stim could be at least in part explained by their opposite effects on contractile protein expression and organization.

(A and B) Representative longitudinal myobundle sections immunostained for (A) F-actin (green; scale bars, 100 m) and (B) SAA (red; scale bars, 50 m). (C and D) Quantification of (C) projected myotube length (n > 350 myotubes from three donors per group) and (D) SAA cross-striation frequency (n 9 images from each donor, n 37 images per group). (E) Representative Western blots from a single donor showing expression of the sarcomeric proteins dystrophin, myosin heavy chain (MYH; MF20), SAA, and myosin light chain (MYL; F-5), with glyceraldehyde-3-phosphate dehydrogenase (GAPDH) serving as a loading control. (F to I) Quantification of Western blots averaged for three donors with protein abundance normalized to GAPDH and shown relative to IFN- group. *P < 0.05 versus IFN-, #P < 0.05 versus IFN-+, **P < 0.05. NS, nonsignificant. Data are presented as means SEM.

To determine whether IFN-induced force deficit may be additionally caused by impaired calcium handling, we measured myobundle Ca2+ transients using Fluo-8 AM dye (Fig. 4, A to D, and movie S2). IFN- treatment reduced calcium transient amplitude to ~60% of that in the untreated myobundles (Fig. 4E and table S1). E-stim caused a significant increase in Ca2+ transient amplitude in both untreated and IFN-treated myobundles, consistent with changes in contractile force generation (Fig. 1G). To further obtain molecular insights in altered calcium handling in myobundles, we analyzed expression of RYR1 and CASQ1, proteins involved in sarcoplasmic reticulum Ca2+ release and buffering (Fig. 4F). Consistent with the observed decrease in Ca2+ transient amplitude, RYR1 and CASQ1 expressions were significantly reduced following IFN- treatment (Fig. 4, G and H). E-stim prevented the IFN-induced effects, significantly increasing the expression of RYR1 and CASQ1 to and beyond the control levels in untreated myobundles (Fig. 4, G and H). Collectively, these results revealed that IFN-induced weakness in myobundles was caused by deficits in both myotube structure and calcium handling.

(A to D) Representative peak Fluo-8 AM fluorescence intensity during E-stim (1 and 20 Hz) showing amplitudes of Ca2+ transients; scale bars, 500 m. (E) Quantified amplitudes (F/F) of electrically stimulated Ca2+ transients based on Fluo-8 AM recording (n = 8 myobundles from one donor per group). (F) Representative Western blots for RYR1, CASQ1, and GAPDH (loading control). (G and H) Quantified (G) RYR1 and (H) CASQ1 protein expressions normalized to that of GAPDH and shown relative to IFN- group (n = 6 samples from three donors per group). (I) Representative Western blots for pSTAT1, STAT1, and GAPDH (loading control). (J to L) Quantified (J) STAT1, (K) pSTAT1, and (L) pSTAT1/STAT1 protein expressions normalized to that of GAPDH and shown relative to the IFN-+ group (n = 6 samples from three donors per group). *P < 0.05 versus IFN-, #P < 0.05 versus IFN-+. NS, nonsignificant. Data are presented as means SEM.

Regarding the known roles of IFN- in secretion and sensing of various cytokines, we quantified secretome of myobundles cultured in serum-free media in response to IFN- and/or E-stim using a multiplex beadbased assay (fig. S5). We found that application of IFN- increased myobundle secretion of several proinflammatory cytokines, namely, IL-7, IL-12p70, CX3CL1, IL-18, and monocyte chemoattractant protein-1 (MCP-1), and reduced myobundle secretion of IL-8 and leukemia inhibitory factor (LIF). E-stim altered myobundle secretome consistent with a report on human myotubes electrically stimulated short-term in serum-containing media (36). The cotreatment of myobundles with E-stim and IFN- partially or fully reversed the IFN-induced effects on IL-12p70, IL-18, IL-8, and LIF and had no effects on MCP-1 or CX3CL1 secretion. Application of E-stim increased the myobundle secretion of IL-6 to a similar level with and without IFN- application, consistent with previous findings in mice suggesting that IFN-induced muscle wasting is IL-6 independent (9).

Previous studies have suggested that proinflammatory effects of IFN- in muscle result from up-regulation of the JAK/STAT pathway (37). We thus measured the protein expression of total STAT1 and phosphorylated STAT1 (pSTAT1) and assessed the degree of STAT1 activation defined as pSTAT1/STAT1 (Fig. 4, I to L). We found that IFN- treatment increased both the total STAT1 and pSTAT1 levels as well as the ratio of pSTAT1/STAT1. While E-stim had no significant effects on STAT1 or pSTAT1 levels in the absence of IFN-, it significantly attenuated IFN-induced pSTAT1 increase without affecting the levels of STAT1 (Fig. 4, I to L). Together, E-stim partially (~50%) attenuated the IFN-induced up-regulation of the JAK/STAT1 pathway in myobundles, which, in addition to direct effects of E-stim on muscle hypertrophy and strengthening, represents a novel, independent mechanism for the beneficial anti-inflammatory effects of exercise on muscle weakness and wasting induced by IFN-.

Beneficial effects of E-stim in conjunction with its partial down-regulation of JAK/STAT1 signaling prompted us to test in an independent set of experiments whether the direct inhibition of JAK/STAT pathway by Food and Drug Administration (FDA)approved small-molecule inhibitors tofacitinib (Tofa, blocker of JAK1/2/3) and baricitinib (Bari, blocker of JAK1/2) can prevent structural and functional deficits induced by IFN- (Fig. 5A). We found that 8-day treatment with either inhibitor (at a clinically relevant dose of 500 nM) had no adverse effects on myobundle morphology and function. On the other hand, the same dose of the inhibitors fully prevented the deteriorating effects of IFN- on myobundle contractile force and kinetics in all of the donors (Fig. 5B and fig. S6, A to F), as well as blocked the IFN-induced increase in myobundle CSA (Fig. 5C) and decrease in myotube size, length, and abundance of cross-striations (Fig. 5, D to F, and fig. S7, A to C). The exact numerical results of functional measurements are presented in table S2. We next investigated whether JAK/STAT inhibitors prevented the IFN-induced changes in myobundle contractile protein expression and calcium handling. Treatment of nave myobundles with the inhibitors had no apparent effects on contractile or calcium handling protein expression or generation of Ca2+ transients (Fig. 6, A to F, and fig. S7, D and E). Coapplication of JAK/STAT inhibitors with IFN- fully prevented the IFN-induced decrease in expression of MYL and MYH (Fig. 6, A to C), Ca2+ transient amplitude (Fig. 6D), and RYR1 and CASQ1 expression (Fig. 6, E and F), without any changes in dystrophin or SAA levels (fig. S7, D and E).

(A) Schematic overview of myobundle culture, treatment, and characterization. Human primary myoblasts were expanded in 2D culture and mixed with hydrogel to form 3D myobundles, which were cultured in GM for 4 days, then in DM for 6 days, after which JAK inhibitors (JAKi) tofacitinib (Tofa, 500 nM) or baricitinib (Bari, 500 nM) was applied for additional 8 days, the last 7 of which in the presence or absence of IFN-. (B) Contractile force amplitude per myobundle CSA (specific force, n = 11 to 15 myobundles from three donors per group). (C) Quantified CSA of myobundles. (D) Quantified myotube diameter from myobundle cross sections (n 720 myotubes from three donors per group). (E and F) Quantified (E) projected myotube length (n > 150 myotubes from two donors per group) and (F) SAA cross-striation frequency (n 30 images from three donors per group) from longitudinal sections of myobundles. *P < 0.05 versus IFN-, #P < 0.05 versus IFN-+. NS, nonsignificant. Data are presented as means SEM.

(A) Representative Western blots from a single donor showing expression of dystrophin, MYH (MF20), SAA, and MYL (F-5), with GAPDH serving as a protein loading control. Tofa + I: tofacitinib + IFN-, Bari + I: baricitinib + IFN-. (B and C) Quantifications of Western blots for (B) MYL and (C) MYH averaged for three donors with protein abundance normalized to GAPDH and shown relative to IFN- group (n = 6 samples from three donors per group). No difference in dystrophin or SAA expression was observed. (D) Quantified amplitudes of electrically stimulated (1 and 20 Hz) Ca2+ transients (n = 8 myobundles from one donor per group). (E) Representative Western blots for RYR1, CASQ1, and GAPDH. (F) Quantified RYR1 and CASQ1 protein expression normalized to that of GAPDH and shown relative to the IFN- group (n = 6 samples from three donors per group). (G) Representative Western blots for pSTAT1, STAT1, and GAPDH. (H and I) Quantified (H) pSTAT1 and (I) pSTAT1/STAT1 protein expression normalized to that of GAPDH and shown relative to the IFN-+ group (n = 3 samples from three donors per group). *P < 0.05 versus IFN-, #P < 0.05 versus IFN-+; **P < 0.05. NS, nonsignificant. Data are presented as means SEM.

Because JAK/STAT inhibitors completely prevented the inflammatory action of IFN- on myobundles, we assessed their effects on STAT1 expression and activity by Western blotting. The treatment of nave myobundles with tofacitinib did not affect the expression of STAT1 or pSTAT1, while baricitinib moderately increased the expression of STAT1 but not pSTAT1 (Fig. 6, G to I, and fig. S7F), consistent with previous studies (38). Application of the inhibitors to IFN-treated myobundles significantly decreased pSTAT1 expression to near-nave levels (Fig. 6, G and H), without altering the IFN-induced increase in STAT1 expression (fig. S7F). Consequently, the IFN-induced increase in STAT1 activity (pSTAT1/STAT1) was fully prevented by coapplication of the inhibitors, explaining their strong protective effects on myobundle function. Together, these results established that the adverse effect of IFN- on human myobundles was predominantly mediated via up-regulation of JAK/STAT1 signaling rather than changes in alternative signaling pathways (39).

Here, we used a human myobundle system to model inflammation-induced muscle weakness and explore anti-inflammatory mechanisms of muscle exercise. Previously, we have shown that myobundles represent a unique personalized in vitro platform to study not only structural and biochemical but also metabolic and contractile responses of human skeletal muscle to a diverse set of pathological and physiologic inputs, including exercise (2224, 40). In this study, we sought to explore how human skeletal muscle strength and structure are affected by IFN-, a prototypical cytokine elevated in various inflammatory diseases (18, 41, 42). Consistent with studies in mice (13, 14), 7-day treatment of human myobundles with IFN- (20 ng/ml) induced muscle weakness (i.e., weaker and slower contractions) that was associated with significant myofiber atrophy and disarray, reduced expression of contractile and calcium handling proteins, and altered cytokine expression. These results were fully reproduced in myobundles derived from three independent donors. When coapplied with IFN-, a 7-day intermittent exercise-mimetic E-stim had pronounced protective effects on myobundles. Beside its well-established hypertrophic and strengthening effects (24, 43), the exercise-mimetic E-stim (24) partially reduced IFN-induced STAT1 up-regulation, established using selective JAK/STAT inhibitors to be the dominant proinflammatory mediator of IFN- action in myobundles. To our knowledge, this is the first study to explore direct and specific effects of IFN- on human skeletal muscle function and to demonstrate the existence of a novel myofiber-autonomous, anti-inflammatory mechanism of muscle exercise involving the JAK/STAT1 pathway (fig. S8). We anticipate the future use of myobundle platform to study human inflammatory disease and anti-inflammatory therapies.

Skeletal muscle is an important secretory organ in the body that both responds to systemic endocrine signals and can function as an active regulator of immune and inflammatory response. Physical exercise can alter the cytokine secretion of skeletal muscle and other tissues, which in turn can promote muscle repair after injury (44), inhibit muscle atrophy (26), and potentially act as an anti-inflammatory therapy in chronic inflammatory diseases (27). While the anti-inflammatory effect of exercising muscle fibers has been mainly attributed to their paracrine cross-talk with proinflammatory nonmuscle cells (29), the use of myobundles devoid of immune or fat cells in this study allowed us to unveil muscle-autonomous inhibitory effects of exercise on JAK/STAT1 signaling, a mediator of muscle inflammation caused by IFN-. This exercise-induced down-regulation of pSTAT1 could be autocrine mediated via the partial normalization of IFN-induced changes in myobundle secretome; however, pleotropic effects of muscle exercise on several intracellular signaling pathways (45) and their potential cross-talk with JAK/STAT signaling (39, 46, 47) warrant future investigations. Our studies also confirmed that the FDA-approved small-molecule JAK/STAT inhibitors tofacitinib and baricitinib, prescribed for rheumatoid arthritis, fully prevented IFN-induced muscle wasting at a nontoxic, therapeutically relevant dose (32), although their biochemical mechanisms of action may vary based on the somewhat differing effects on STAT1 and pSTAT1 expression.

In summary, we have applied an in vitro 3D myobundle model to investigate the interplay between IFN- and exercise-mimetic stimulation in inflammatory response of human skeletal muscle. We show that chronic application of IFN- induces myobundle weakness via up-regulation of JAK/STAT1 signaling pathway, which can be partially prevented by exercise-mimetic stimulation and fully prevented by treatment with clinically approved JAK/STAT inhibitors. We envision that incorporation of additional human nonmuscle cells in the myobundle platform [akin to use of macrophages in a rat skeletal muscle model (40)] multiplexed with application of various inflammatory cytokines (18), exercise-mimetic regimes (24), and candidate therapeutics will allow comprehensive mechanistic studies of human muscle inflammation, anti-inflammatory roles of exercise, and discovery of effective therapies for muscle wasting.

Human skeletal muscle samples were obtained from three donors (two females and one male, age 12 to 18) through standard needle biopsy or surgical waste from three donors with informed consent under Duke University Institutional Review Boardapproved protocols (Pro00048509 and Pro00012628). Muscle tissue was minced using sharp scissors and enzymatically digested with 0.05% trypsin for 30 min at 37C. Isolated cells were collected by centrifugation and resuspended in growth media [GM; low-glucose Dulbeccos modified Eagles medium (DMEM) (Thermo Fisher Scientific, D6046) supplemented with 10% fetal bovine serum (FBS) (Hyclone, SH30071.03), fetuin (50 g/ml) from FBS (Sigma-Aldrich, F2379), recombinant human epidermal growth factor (EGF) (PeproTech, AF-100-15), dexamethasone (Sigma-Aldrich, F2379), and penicillin G (100 U/ml, Thermo Fisher Scientific)] and preplated for 2 hours to reduce the number of fibroblasts. After preplating, the cells were transferred onto to 1% Matrigel (BD Biosciences)coated flasks, cultured in GM, and expanded by passaging after reaching 70% confluence. At passage 4 or 5, cells were detached, counted, and used to fabricate myobundles.

Human myobundles were fabricated as described previously (23, 24). Briefly, polydimethylsiloxane (PDMS) molds made to fit in a well of a 12-well plate were casted from 3D-machined Teflon masters containing two semicylindrical wells (7 mm long, 2-mm diameter for culture of two myobundles), sterilized in 70% ethanol, air dried in a tissue culture hood, and coated with 0.2% (w/v) Pluronic F-127 (Invitrogen) for at least 1 hour at room temperature to prevent tissue adhesion. Laser-cut Cerex frames (9 9 mm2, 1-mm-wide rim) were sterilized and placed within the air-dried PDMS molds. The frame provided an anchor surface for myobundle attachment and functioned as a mechanical guide during cell-mediated hydrogel compaction, resulting in uniaxial cell alignment (48). Expanded myogenic cells were dissociated using 0.025% trypsin-EDTA and encapsulated in a hydrogel solution at 1.5 107 cells/ml. For a single myobundle, the cell/hydrogel solution contained 10 l of bovine fibrinogen (20 mg/ml) in phosphate-buffered saline (PBS) (Sigma-Aldrich), 10 l of Matrigel (Corning), 2 l of bovine thrombin (50 U/ml) in 0.1% bovine serum albumin (BSA) in PBS (Sigma-Aldrich), and 28.2 l of GM with 0.75 106 cells, prepared on ice, which was mixed thoroughly and immediately pipetted into one well of the PDMS mold. Cell/hydrogel mixture in PDMS molds was polymerized at 37C for 30 min, and thereafter, myobundles were cultured on a rocker. After 2 days, frames with myobundles were removed from the molds and left to float in culture media. GM was used for the first 4 days of culture, after which it was replaced by differentiation medium [DM, low glucose DMEM (Thermo Fisher Scientific) supplemented with N2 supplement (100, Thermo Fisher Scientific), penicillin G (100 U/ml, Thermo Fisher Scientific)], with media changes performed daily. Media were supplemented with 6-aminocaproic acid (ACA; Sigma-Aldrich) to reduce fibrinolysis (1.5 mg/ml in GM. 2 mg/ml in DM).

For inflammation experiments, starting on differentiation day 7 (culture day 11), myobundles were treated for an additional 7 days with IFN- (20 ng/ml) [PeproTech, 300-02, in BSA final (0.5 g/ml)] freshly added during each media change. Control group was supplemented with media only. For E-stim experiments, we used custom-made PDMS chambers containing two parallel carbon electrodes, as previously described (24, 49). Myobundles were placed in the chamber, and E-stim was applied from differentiation days 7 to 14 either without or with IFN- treatment using a D330 MultiStim system (Digitimer Ltd.) controlled by a custom-made LabVIEW program. The applied intermittent regime of stimulation consisted of 1-hour stimulation cycles (bipolar 2 ms, 70-mA constant-current impulses applied in 0.5-s-long 10-Hz pulse trains delivered every 5 s) separated by 7-hour rests (Fig. 1B) (24). For experiments with JAK/STAT inhibitors, tofacitinib (in water; Sigma-Aldrich, PZ0017) or baricitinib (in 0.01% DMSO final; Sigma-Aldrich, G-5743,) was added at 500 nM concentration to myobundles starting on differentiation day 6, 1 day before IFN- treatment, and continued to be freshly applied (with or without IFN-) for the following 8 days during the daily media changes.

Contractile force generation and passive tension in myobundles were measured using a previously described custom-made force measurement apparatus (22). Briefly, single myobundles were transferred to the force measurement apparatus and immersed in DMEM media at 37C. One end of the myobundle was attached to a fixed PDMS block, and the other to a movable PDMS float connected to a force transducer mounted on a computer-controlled motorized linear actuator (Thorlabs, Newton, NJ). The sides of the frame were cut to allow force measurements. Using the linear actuator, the myobundle was stretched to 100, 108, and 116% of their original culture length, and twitch contractions were recorded in response to 10-ms electrical pulses applied by platinum electrodes at 1-Hz stimulation rate. At 16% stretch, 1-s-long stimulation at 20 Hz was applied to record tetanic contractile force. Force traces were analyzed for peak twitch and tetanus force, passive tension, time-to-peak twitch, and half-relaxation time using a custom MATLAB program (22, 40).

Myobundles were fixed in 2% (v/v) paraformaldehyde (Electron Microscopy Sciences) in PBS overnight at 4C with rocking. Fixed samples were submerged in optimal cutting temperature compound (Electron Microscopy Sciences) and snap frozen in liquid nitrogen. Constructs were sectioned (10 m thick) parallel (longitudinal) and perpendicular (cross) to the long axis of the bundles using a cryostat (LEICA CM1950) and were mounted onto glass slides. Before staining, sections were incubated in a blocking solution containing 5% chick serum and 0.1% Triton X-100 in PBS overnight at 4C. Primary antibodies were applied in blocking solution overnight at 4C. Secondary antibodies were applied overnight at 4C. Antibody information and dilutions are listed in table S3. Immunostained samples were mounted with ProLong Glass Antifade reagent (Thermo Fisher Scientific, P36984). Fluorescence images were acquired using an Andor Dragonfly spinning disk confocal microscope at 10 to 40 magnification and analyzed by ImageJ.

Images of myobundle longitudinal sections stained for F-actin and SAA were used for the measurement of myotube organization. Projected length of clearly distinguishable F-actinlabeled myotubes was determined manually (fig. S3, A and B). Percentage of cross-striated myotubes was assessed by manually counting the cross-striated myotubes labeled by SAA staining divided by the total number of myotubes determined from F-actin costaining of the same section.

Media conditioned by myobundles from three donors were collected between days 3 and 4 of E-stim for the E-stim groups and on equivalent culture days (between days 14 and 15) for the groups without E-stim, then frozen, and used for the secretome analysis on the same plate upon thawing. Select cytokine concentrations were measured using a custom-designed human magnetic 18-plex panel for the Luminex platform (Thermo Fisher Scientific, Waltham, MA) by the Immunology Core at Duke University following the manufacturers instructions.

Protein was isolated from three to four myobundles per experimental sample in ice-cold radioimmunoprecipitation assay lysis and extraction buffer in the presence of protease inhibitor (Sigma-Aldrich) and phosphatase inhibitor cocktail (Roche), as previously described (24). Protein concentration was determined using a bicinchoninic acid assay (Thermo Fisher Scientific). Western blots were performed using Bio-Rad Mini-PROTEAN 4 to 15% gradient gels with proteins transferred using a Bio-Rad Mini Trans-Blot Cell. After blocking with 5% milk or 5% BSA in tris-buffered saline with 0.1% Tween 20 at room temperature for 1 hour, primary antibodies diluted in the same blocking solution were incubated with the membrane at 4C overnight. Horseradish peroxidaseconjugated anti-mouse and anti-rabbit secondary antibodies were applied for 1 hour at room temperature. Antibody information and dilutions are listed in table S3. Protein detection was performed using either SuperSignal West Pico PLUS or Femto Maximum ECL chemiluminescence substrates (Thermo Fisher Scientific). Images were acquired using a Bio-Rad ChemiDoc imaging system and analyzed with ImageJ.

For Ca2+ transient measurements, myobundles were incubated with 50 M of calcium-sensitive dye Fluo-8 AM (Abcam, ab142773) in DM in an incubator for 1 hour while rocking, followed by washing in dye-free media for 30 min. Electrically induced Ca2+ transients were recorded as previously described (24, 40). Myobundles were transferred into a glass-bottom live-imaging chamber with Tyrodes solution warmed at 37C in a heated live-imaging chamber. Fluorescence images were acquired at 4 magnification on a Nikon microscope using a high speed EMCCD (electron multiplying charge-coupled device) camera (Andor iXon 860) and Andor Solis software. Ca2+ transient amplitudes were calculated as the maximum relative change in fluorescence signal, F/F = (Peak Trough)/(Trough Background).

Experimental data are presented as means SEM. Statistical significances between groups were verified by one-way analysis of variance (ANOVA) with Tukeys post hoc test or as described in the figure captions, using GraphPad Prism (GraphPad Software). P < 0.05 was considered statistically significant. Sample sizes for in vitro experiments were determined based on variance of previously reported measurements. All immunostaining images and movies shown are representative of similar results from at least three independent experiments.

Acknowledgments: We thank A. Khodabukus for technical advice and K. Huang for advice on writing the manuscript. Funding: Research reported in this study was supported by the NIH under grants UH3TR002142, U01EB028901, and R01AR070543. The content of the manuscript is solely the responsibility of the authors and does not necessarily represent the official views of the NIH. Author contributions: N.B., L.R., and Z.C. conceived the idea and designed the experiments; Z.C., L.R., B.L., and R.-Z.Z. performed the experiments and analyzed the data; Z.C. and N.B. wrote the manuscript. 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:
Exercise mimetics and JAK inhibition attenuate IFN-induced wasting in engineered human skeletal muscle - Science Advances

Can Science Save the Northern White Rhino? – Freethink

The northern white rhino reproduces like most mammals: a male and a female have sex, the female gets pregnant, and about 18 months later, a new rhino calf is born.

But right now, there aren't any male northern white rhinos the last one, Sudan, died in 2018 and just two females remain.

In the past, that would mean the species' total extinction was just on the horizon.

Today, though, teams of researchers are exploring three different plans to revive the northern white rhino and one involves a controversial technique that could also revolutionize human reproduction.

The first plan for saving the northern white rhino is straightforward, science-wise: in vitro fertilization (IVF).

Prior to his death, Sudan lived at Ol Pejeta Conservancy, a protected wildlife area in Kenya, along with the only remaining females of his species, Najin and her daughter Fatu.

When Sudan and four other males were still alive, Ol Pejeta researchers collected samples of their sperm. They've since used some of that sperm to fertilize eggs they retrieved from Najin and Fatu in 2019.

They hope to inseminate the potential mothers before 2022.

This has resulted in five viable embryos.

Both Fatu and Najin have known reproductive issues that would prevent them from being able to carry a calf to term, so researchers plan to use female southern white rhinos a close relative of the northern white rhino, with numbers in the ten thousands as surrogates.

Their hope is to complete the insemination of the potential mothers before 2022.

However, no one knows for sure whether the pregnancy will take, as rhino reproduction is complicated. The first southern white rhino conceived through artificial insemination was only just born in July 2019, and no one has been able to produce one through IVF yet.

Southern white rhinos are also at the center of another plan to save their close relatives from extinction.

The procedure researchers developed to extract eggs from Fatu and Najin required the use of a risky anesthetic, so before subjecting the pair to it, they tested the technique on 12 southern white rhinos.

They then used sperm from northern white rhinos to fertilize some of the eggs they collected.

If any calves were born from those hybrid embryos, they'd be half northern white rhino and half southern, which would keep the former species somewhat alive.

Still, Fatu and Najin would be the last fully northern white rhinos, and their deaths would bring about the species' permanent extinction unless scientists go to Plan C, a very experimental (and sometimes controversial) technique called in vitro gametogenesis (IVG).

In addition to collecting sperm and eggs from the last northern white rhinos, researchers have also collected and frozen tissue samples from about a dozen members of the species.

In 2011, a group at Scripps Research Institute in California proved it was possible to create induced pluripotent stem cells (iPSC) from this rhino tissue. These cells can be prompted to grow into any type of specialized cell under the right conditions including reproductive cells.

In theory, researchers could prompt northern white rhino cells to develop into sperm and eggs, fertilize the eggs with the sperm, and then implant the embryos into surrogates (like the southern white rhinos).

Researchers have actually used IVG like this to successfully impregnate mice.

If it works in rhinos or other animals, it could bring whole species back from extinction (assuming you had enough tissue samples and a surrogate mother species).

If scientists can make it work in humans, it would completely change the parameters for reproduction. Single people could have babies related only to them, creating their own eggs and sperm, and same-sex couples could have tots that are related to them both (though two men would need to find a surrogate to carry the baby).

Again, IVG has only been shown in mice so far, so the use of such an experimental technique in rhinos would likely be years down the line (and humans even further).

However, it does offer hope that the northern white rhino species doesn't have to die with Fatu and Najin, even after their last egg is gone.

We'd love to hear from you! If you have a comment about this article or if you have a tip for a future Freethink story, please email us at [emailprotected].

Link:
Can Science Save the Northern White Rhino? - Freethink

Induced Pluripotent Stem Cells Market to Grow with an Impressive CAGR – Farming Sector

The global market for induced pluripotent stem cells (iPSCs) reached $2.1 billion in 2016. The market should reach $3.6 billion in 2021, increasing at a compound annual growth rate (CAGR) of 11.6% from 2016 through 2021.

Report Scope:

This study is focused on the market side of iPSCs rather than its technical side. Different market segments for this emerging market are covered. For example, application-based market segments include academic research, drug development and toxicity testing, and regenerative medicine; product function-based market segments include molecular and cellular engineering, cellular reprogramming, cell culture, cell differentiation and cell analysis; iPSC-derived cell-type-based market segments include cardiomyocytes, hepatocytes, neurons, endothelia cells and other cell types; geography-based market segments include the U.S., Europe, Asia-Pacific and Rest of World. Research and market trends are also analyzed by studying the funding, patent publications and research publications in the field.

Get PDF Sample for the Report:https://www.trendsmarketresearch.com/report/sample/12035

Report Includes:

An overview of the global market for induced pluripotent stem cells. Analyses of global market trends with data from 2015 and 2016, and projections of compound annual growth rates (CAGRs) through 2021. Information on induced pluripotent stem cell research products, defined as all research tools including but not limited to: induced pluripotent stem cells and various differentiated cells derived from induced pluripotent stem cells; various related assays and kits, culture media and medium components, such as serum, growth factors and inhibitors, antibodies, enzymes, and many others that can be applied for the specific purpose of executing induced pluripotent stem cell research. Discussion of important manufacturers, technologies, and factors influencing market demand, such as the driving forces and limiting factors of induced pluripotent stem cell market growth. Profiles of major players in the industry.

Report Summary

Its been over 10 years since the discovery of induced pluripotent stem cell (iPSC) technology. The market has gradually become an important part of the life sciences industry during recent years. Particularly for the past five years, the global market for iPSCs has experienced a rapid growth. The market was estimated at $1.7 billion in 2015 and over $2 billion in 2016, with an average 18% growth. The overall iPSC market is forecast to continue its relatively rapid growth and reach over $3.6 billion in 2021, with an estimated compound annual growth rate (CAGR) of 11.6% from 2016 through 2021.

You can Buy This Report from Here @https://www.trendsmarketresearch.com/checkout/12037/Single

Key Drivers for Market Growth

This report has identified several key drivers for the rapidly growing market: iPSC shold promising hope for therapeutic solutions for diseases without ethical issues. A series of technical breakthroughs were made in recent years for improving cellular reprogramming, differentiation and large-scale production of GMP- grade iPSCs derived cells toward clinical usability. The pharmaceutical industry needs better cell sources such as iPSC-derived functional cells for drug toxicity testing and drug screening. The U.S. government has been encouraging the marketing of stem cells, including iPSCs. The U.S. Food and Drug Administration (FDA) has been authorized to provide orphan drug designations for many of the therapies developed for rare diseases such as Parkinsons and Huntingtons using stem cells. The provisions of grants from organizations, such as the National Institutes of Health (NIH) and the California Institute for Regenerative Medicine (CIRM) have been encouraging for the research institutes to venture into iPSC research. Rapidly growing medical tourism and contract research outsourcing drives the Asia-Pacific stem cell market. Cellular reprogramming, including iPSC technology, was awarded the 2012 Nobel Prize. The first human iPSC clinical trial started in August 2014, and the recent report of the first macular degeneration patient treated with the sheets of retinal pigmented epithelial cells made from iPSCs was encouraging. iPSC technology is developing into a platform for precision and personalized medicine, which is experiencing rapid growth globally. New biotechnologies such as genome editing technology are advancing iPSCs into more and better uses.

This report identifies key revenue segments for the iPSC market from various aspects. The applicationbased segments include the research, drug development and clinical markets; the product functionbased segments include molecular and cellular engineering, cellular reprogramming, cell culture, cell differentiation and cell analysis. The current major revenue segment is the drug development and toxicity testing sector, but the market for regenerative medicine is the fastest growing one. The market for clinical applications is not fully established, but the market for the translational medicine research of iPSC is also growing very quickly.

Read the original here:
Induced Pluripotent Stem Cells Market to Grow with an Impressive CAGR - Farming Sector

Accelerating cell-based therapies by providing safe therapeutic MSC products – BioSpace

TORONTO, Jan. 8, 2021 /CNW/ -panCELLa's intent to develop therapeutic products in the mesenchymal stem cell (MSC) and pancreatic islet space has recently led to the creation of Implant Therapeutics.

Implant Therapeutics, under the guidance of Dr. Mahendra Rao, is engineering iPSC-MSC cells containing panCELLa's FailSafe and induced Allogeneic Cell Tolerance (iACT Stealth Cell) technologies. These iPSC MSC cells are hypo-immunogenic and are an ideal choice for bone, cartilage and tendon replacement strategies combining the advantages of allogeneic and autologous cells as well as allowing them to be used as ex-vivo gene therapy vehicles.

Implant is pleased to announce a definitive cross licensing agreement with RxCell. The collaboration will allow both companies to rapidly move forward in their respective fields with enhanced technology platforms, access to cGMP grade iPSC lines and the ability to generate a wide variety of therapeutic grade products.

"RxCell's hypo-immunogenic cell platform complements our cloaking platform and allows us to develop novel tissue-specific hypo-immunogenic cell lines" said Dr. Andras Nagy, the inventor of the cloaking technology otherwise known as panCELLa's iACT (Stealth Cells). Dr. Zeng added that she was particularly pleased to have access to cGMP grade engineered Master Cell Banks that utilized Sigma/Merck CRISPR-based technology to incorporate the safe harbor technology.

Dr. Rao CEO of Implant Therapeutics added "I believe that the MSC platform developed by the two companies combines the proven power of MSC with the engineering expertise of panCELLa and this will make RxCell and Implant leaders in their respective fields."

About Implant Therapeutics

Implant provides hypoimmunogenic and safe harbor engineered IPSC derived cellsin order to deliver the ultimate therapeutic MSC products. To learn more, visit https://www.implant-rx.com/

About panCELLa

Founded in 2015, panCELLa is a privately-held early-stage biotechnology firm based on the innovative technology developed in Dr. Andras Nagy's lab at the Sinai Health System (SHS). panCELLa has created platforms that allow for the development of safe, universal, "off-the-shelf" cell lines. To learn more, visit https://pancella.com.

About RxCell

RxCell is a biotechnology company focused on therapeutic applications of induced pluripotent stem cells (iPSC). We have developed several therapeutic grade iPSC line seed banks and are in the process of manufacturing a large Master Cell Bank to support our activities. To learn more, visit https://www.rxcellinc.com/

SOURCE panCELLa Inc.

Read this article:
Accelerating cell-based therapies by providing safe therapeutic MSC products - BioSpace