Search Results for: stem cell harvest adipose

Stem Cell Treatment Centers – Indiana Stem Cell | Call Today

Posted: June 23, 2021 at 1:52 am

At the Indiana Stem Cell Treatment Center, we provide stem cell therapy care for people suffering from diseases that may be alleviated by access to adult stem cell based regenerative treatment. The Center utilizes a fat transfer surgical technology to isolate and implant the patients own stem cells from a small quantity of fat harvested by liposuction on the same day. Stem cell therapy patients are evaluated by a respective member of our multi-specialty expert panel of Board Certified physicians representing many medical fields. The Indiana Stem Cell Treatment Center emphasizes quality and is highly committed to clinical research and the advancement of regenerative medicine. When it comes to stem cell therapy centers we always put the patients needs first

Founded in 2010 for the investigational use of stem cells deployments for degenerative conditions, the source of the cells is actually stromal vascular fraction, which is a protein rich segment of processed adipose tissue. Stromal vascular fraction contains a mononuclear cell line (predominantly autologous mesenchymal stem cells), macrophage cells, endothelial cells, red blood cells, and important growth factors that turn on the stem cells and promote their activity. We have high numbers of viable cells and we are trying to learn which diseases respond best and which deployment methods are most effective. We are growing and continue to use our surgical methods to deploy SVF for various degenerative conditions. We employ a clinical research coordinator to protect our valuable data and our vision is to perfect our treatments and ultimately teach them to other physicians around the world.

Visit link:
Stem Cell Treatment Centers - Indiana Stem Cell | Call Today

Posted in Indiana Stem Cells | Comments Off on Stem Cell Treatment Centers – Indiana Stem Cell | Call Today

Global Cell Harvesting Market to Reach US$381,4 Million by the Year 2027 – Salamanca Press

Posted: November 30, 2020 at 2:59 pm

NEW YORK, Nov. 25, 2020 /PRNewswire/ --Amid the COVID-19 crisis, the global market for Cell Harvesting estimated at US$233.2 Million in the year 2020, is projected to reach a revised size of US$381.4 Million by 2027, growing at a CAGR of 7.3% over the period 2020-2027.Manual, one of the segments analyzed in the report, is projected to grow at a 7.9% CAGR to reach US$284.4 Million by the end of the analysis period. After an early analysis of the business implications of the pandemic and its induced economic crisis, growth in the Automated segment is readjusted to a revised 5.6% CAGR for the next 7-year period. This segment currently accounts for a 28.3% share of the global Cell Harvesting market.

Read the full report: https://www.reportlinker.com/p05798117/?utm_source=PRN

The U.S. Accounts for Over 30.9% of Global Market Size in 2020, While China is Forecast to Grow at a 10.4% CAGR for the Period of 2020-2027

The Cell Harvesting market in the U.S. is estimated at US$72 Million in the year 2020. The country currently accounts for a 30.86% share in the global market. China, the world second largest economy, is forecast to reach an estimated market size of US$34.9 Million in the year 2027 trailing a CAGR of 10.4% through 2027. Among the other noteworthy geographic markets are Japan and Canada, each forecast to grow at 6.1% and 7% respectively over the 2020-2027 period. Within Europe, Germany is forecast to grow at approximately 6.6% CAGR while Rest of European market (as defined in the study) will reach US$34.9 Million by the year 2027.We bring years of research experience to this 5th edition of our report. The 226-page report presents concise insights into how the pandemic has impacted production and the buy side for 2020 and 2021. A short-term phased recovery by key geography is also addressed.

Competitors identified in this market include, among others,

Read the full report: https://www.reportlinker.com/p05798117/?utm_source=PRN

I. INTRODUCTION, METHODOLOGY & REPORT SCOPE I-1

II. EXECUTIVE SUMMARY II-1

1. MARKET OVERVIEW II-1Cell Harvesting - A Prelude II-1Impact of Covid-19 and a Looming Global Recession II-1With Stem Cells Holding Potential to Emerge as Savior forHealthcare System Struggling with COVID-19 Crisis, Demand forCell Harvesting to Grow II-1Select Clinical Trials in Progress for MSCs in the Treatment ofCOVID-19 II-2Lack of Antiviral Therapy Brings Spotlight on MSCs as PotentialOption to Treat Severe Cases of COVID-19 II-3Stem Cells Garner Significant Attention amid COVID-19 Crisis II-3Growing R&D Investments & Rising Incidence of Chronic Diseasesto Drive the Global Cell Harvesting Market over the Long-term II-3US Dominates the Global Market, Asia-Pacific to ExperienceLucrative Growth Rate II-4Biopharmaceutical & Biotechnology Firms to Remain Key End-User II-4Remarkable Progress in Stem Cell Research Unleashes UnlimitedAvenues for Regenerative Medicine and Drug Development II-4Drug Development II-5Therapeutic Potential II-5

2. FOCUS ON SELECT PLAYERS II-6Recent Market Activity II-7Innovations and Advancements II-7

3. MARKET TRENDS & DRIVERS II-8Development of Regenerative Medicine Accelerates Demand forCell Harvesting II-8The Use of Mesenchymal Stem Cells in Regenerative Medicine toDrive the Cell Harvesting Market II-8Rise in Volume of Orthopedic Procedures Boosts Prospects forStem Cell, Driving the Cell Harvesting II-9Exhibit 1: Global Orthopedic Surgical Procedure Volume (2010-2020) (in Million) II-11Increasing Demand for Stem Cell Based Bone Grafts: PromisingGrowth Ahead for Cell Harvesting II-11Spectacular Advances in Stem Cell R&D Open New Horizons forRegenerative Medicine II-12Exhibit 2: Global Regenerative Medicines Market by Category(2019): Percentage Breakdown for Biomaterials, Stem CellTherapies and Tissue Engineering II-13Stem Cell Transplants Drive the Demand for Cell Harvesting II-13Rise in Number of Hematopoietic Stem Cell TransplantationProcedures Propels Market Expansion II-15Growing Incidence of Chronic Diseases to Boost the Demand forCell Harvesting II-16Exhibit 3: Global Cancer Incidence: Number of New Cancer Casesin Million for the Years 2018, 2020, 2025, 2030, 2035 and 2040 II-17Exhibit 4: Global Number of New Cancer Cases and Cancer-relatedDeaths by Cancer Site for 2018 II-18Exhibit 5: Number of New Cancer Cases and Deaths (in Million)by Region for 2018 II-19Exhibit 6: Fatalities by Heart Conditions: Estimated PercentageBreakdown for Cardiovascular Disease, Ischemic Heart Disease,Stroke, and Others II-19Exhibit 7: Rising Diabetes Prevalence Presents Opportunity forCell Harvesting: Number of Adults (20-79) with Diabetes (inMillions) by Region for 2017 and 2045 II-20Ageing Demographics to Drive Demand for Stem Cell Banking II-20Global Aging Population Statistics - Opportunity Indicators II-21Exhibit 8: Expanding Elderly Population Worldwide: Breakdown ofNumber of People Aged 65+ Years in Million by GeographicRegion for the Years 2019 and 2030 II-21Exhibit 9: Life Expectancy for Select Countries in Number ofYears: 2019 II-22High Cell Density as Major Bottleneck Leads to Innovative CellHarvesting Methods II-22Advanced Harvesting Systems to Overcome Centrifugation Issues II-23Sophisticated Filters for Filtration Challenges II-23Innovations in Closed Systems Boost Efficiency & Productivityof Cell Harvesting II-23Enhanced Harvesting and Separation of Micro-Carrier Beads II-24

4. GLOBAL MARKET PERSPECTIVE II-25Table 1: World Current & Future Analysis for Cell Harvesting byGeographic Region - USA, Canada, Japan, China, Europe,Asia-Pacific and Rest of World Markets - Independent Analysisof Annual Sales in US$ Thousand for Years 2020 through 2027 II-25

Table 2: World Historic Review for Cell Harvesting byGeographic Region - USA, Canada, Japan, China, Europe,Asia-Pacific and Rest of World Markets - Independent Analysisof Annual Sales in US$ Thousand for Years 2012 through 2019 II-26

Table 3: World 15-Year Perspective for Cell Harvesting byGeographic Region - Percentage Breakdown of Value Sales forUSA, Canada, Japan, China, Europe, Asia-Pacific and Rest ofWorld Markets for Years 2012, 2020 & 2027 II-27

Table 4: World Current & Future Analysis for Manual byGeographic Region - USA, Canada, Japan, China, Europe,Asia-Pacific and Rest of World Markets - Independent Analysisof Annual Sales in US$ Thousand for Years 2020 through 2027 II-28

Table 5: World Historic Review for Manual by Geographic Region- USA, Canada, Japan, China, Europe, Asia-Pacific and Rest ofWorld Markets - Independent Analysis of Annual Sales in US$Thousand for Years 2012 through 2019 II-29

Table 6: World 15-Year Perspective for Manual by GeographicRegion - Percentage Breakdown of Value Sales for USA, Canada,Japan, China, Europe, Asia-Pacific and Rest of World for Years2012, 2020 & 2027 II-30

Table 7: World Current & Future Analysis for Automated byGeographic Region - USA, Canada, Japan, China, Europe,Asia-Pacific and Rest of World Markets - Independent Analysisof Annual Sales in US$ Thousand for Years 2020 through 2027 II-31

Table 8: World Historic Review for Automated by GeographicRegion - USA, Canada, Japan, China, Europe, Asia-Pacific andRest of World Markets - Independent Analysis of Annual Sales inUS$ Thousand for Years 2012 through 2019 II-32

Table 9: World 15-Year Perspective for Automated by GeographicRegion - Percentage Breakdown of Value Sales for USA, Canada,Japan, China, Europe, Asia-Pacific and Rest of World for Years2012, 2020 & 2027 II-33

Table 10: World Current & Future Analysis for Peripheral Bloodby Geographic Region - USA, Canada, Japan, China, Europe,Asia-Pacific and Rest of World Markets - Independent Analysisof Annual Sales in US$ Thousand for Years 2020 through 2027 II-34

Table 11: World Historic Review for Peripheral Blood byGeographic Region - USA, Canada, Japan, China, Europe,Asia-Pacific and Rest of World Markets - Independent Analysisof Annual Sales in US$ Thousand for Years 2012 through 2019 II-35

Table 12: World 15-Year Perspective for Peripheral Blood byGeographic Region - Percentage Breakdown of Value Sales forUSA, Canada, Japan, China, Europe, Asia-Pacific and Rest ofWorld for Years 2012, 2020 & 2027 II-36

Table 13: World Current & Future Analysis for Bone Marrow byGeographic Region - USA, Canada, Japan, China, Europe,Asia-Pacific and Rest of World Markets - Independent Analysisof Annual Sales in US$ Thousand for Years 2020 through 2027 II-37

Table 14: World Historic Review for Bone Marrow by GeographicRegion - USA, Canada, Japan, China, Europe, Asia-Pacific andRest of World Markets - Independent Analysis of Annual Sales inUS$ Thousand for Years 2012 through 2019 II-38

Table 15: World 15-Year Perspective for Bone Marrow byGeographic Region - Percentage Breakdown of Value Sales forUSA, Canada, Japan, China, Europe, Asia-Pacific and Rest ofWorld for Years 2012, 2020 & 2027 II-39

Table 16: World Current & Future Analysis for Umbilical Cord byGeographic Region - USA, Canada, Japan, China, Europe,Asia-Pacific and Rest of World Markets - Independent Analysisof Annual Sales in US$ Thousand for Years 2020 through 2027 II-40

Table 17: World Historic Review for Umbilical Cord byGeographic Region - USA, Canada, Japan, China, Europe,Asia-Pacific and Rest of World Markets - Independent Analysisof Annual Sales in US$ Thousand for Years 2012 through 2019 II-41

Table 18: World 15-Year Perspective for Umbilical Cord byGeographic Region - Percentage Breakdown of Value Sales forUSA, Canada, Japan, China, Europe, Asia-Pacific and Rest ofWorld for Years 2012, 2020 & 2027 II-42

Table 19: World Current & Future Analysis for Adipose Tissue byGeographic Region - USA, Canada, Japan, China, Europe,Asia-Pacific and Rest of World Markets - Independent Analysisof Annual Sales in US$ Thousand for Years 2020 through 2027 II-43

Table 20: World Historic Review for Adipose Tissue byGeographic Region - USA, Canada, Japan, China, Europe,Asia-Pacific and Rest of World Markets - Independent Analysisof Annual Sales in US$ Thousand for Years 2012 through 2019 II-44

Table 21: World 15-Year Perspective for Adipose Tissue byGeographic Region - Percentage Breakdown of Value Sales forUSA, Canada, Japan, China, Europe, Asia-Pacific and Rest ofWorld for Years 2012, 2020 & 2027 II-45

Table 22: World Current & Future Analysis for OtherApplications by Geographic Region - USA, Canada, Japan, China,Europe, Asia-Pacific and Rest of World Markets - IndependentAnalysis of Annual Sales in US$ Thousand for Years 2020 through2027 II-46

Table 23: World Historic Review for Other Applications byGeographic Region - USA, Canada, Japan, China, Europe,Asia-Pacific and Rest of World Markets - Independent Analysisof Annual Sales in US$ Thousand for Years 2012 through 2019 II-47

Table 24: World 15-Year Perspective for Other Applications byGeographic Region - Percentage Breakdown of Value Sales forUSA, Canada, Japan, China, Europe, Asia-Pacific and Rest ofWorld for Years 2012, 2020 & 2027 II-48

Table 25: World Current & Future Analysis for Biotech &Biopharma Companies by Geographic Region - USA, Canada, Japan,China, Europe, Asia-Pacific and Rest of World Markets -Independent Analysis of Annual Sales in US$ Thousand for Years2020 through 2027 II-49

Table 26: World Historic Review for Biotech & BiopharmaCompanies by Geographic Region - USA, Canada, Japan, China,Europe, Asia-Pacific and Rest of World Markets - IndependentAnalysis of Annual Sales in US$ Thousand for Years 2012 through2019 II-50

Table 27: World 15-Year Perspective for Biotech & BiopharmaCompanies by Geographic Region - Percentage Breakdown of ValueSales for USA, Canada, Japan, China, Europe, Asia-Pacific andRest of World for Years 2012, 2020 & 2027 II-51

Table 28: World Current & Future Analysis for ResearchInstitutes by Geographic Region - USA, Canada, Japan, China,Europe, Asia-Pacific and Rest of World Markets - IndependentAnalysis of Annual Sales in US$ Thousand for Years 2020 through2027 II-52

Table 29: World Historic Review for Research Institutes byGeographic Region - USA, Canada, Japan, China, Europe,Asia-Pacific and Rest of World Markets - Independent Analysisof Annual Sales in US$ Thousand for Years 2012 through 2019 II-53

Table 30: World 15-Year Perspective for Research Institutes byGeographic Region - Percentage Breakdown of Value Sales forUSA, Canada, Japan, China, Europe, Asia-Pacific and Rest ofWorld for Years 2012, 2020 & 2027 II-54

Table 31: World Current & Future Analysis for Other End-Uses byGeographic Region - USA, Canada, Japan, China, Europe,Asia-Pacific and Rest of World Markets - Independent Analysisof Annual Sales in US$ Thousand for Years 2020 through 2027 II-55

Table 32: World Historic Review for Other End-Uses byGeographic Region - USA, Canada, Japan, China, Europe,Asia-Pacific and Rest of World Markets - Independent Analysisof Annual Sales in US$ Thousand for Years 2012 through 2019 II-56

Table 33: World 15-Year Perspective for Other End-Uses byGeographic Region - Percentage Breakdown of Value Sales forUSA, Canada, Japan, China, Europe, Asia-Pacific and Rest ofWorld for Years 2012, 2020 & 2027 II-57

III. MARKET ANALYSIS III-1

GEOGRAPHIC MARKET ANALYSIS III-1

UNITED STATES III-1Increasing Research on Stem Cells for Treating COVID-19 todrive the Cell Harvesting Market III-1Rising Investments in Stem Cell-based Research Favors CellHarvesting Market III-1Exhibit 10: Stem Cell Research Funding in the US (in US$Million) for the Years 2011 through 2017 III-2A Strong Regenerative Medicine Market Drives Cell HarvestingDemand III-2Arthritis III-3Exhibit 11: Percentage of Population Diagnosed with Arthritisby Age Group III-3Rapidly Ageing Population: A Major Driving Demand for CellHarvesting Market III-4Exhibit 12: North American Elderly Population by Age Group(1975-2050) III-4Increasing Incidence of Chronic Diseases Drives Focus onto CellHarvesting III-5Exhibit 13: CVD in the US: Cardiovascular Disease* Prevalencein Adults by Gender & Age Group III-5Rising Cancer Cases Spur Growth in Cell Harvesting Market III-5Exhibit 14: Estimated Number of New Cancer Cases and Deaths inthe US (2019) III-6Exhibit 15: Estimated New Cases of Blood Cancers in the US(2020) - Lymphoma, Leukemia, Myeloma III-7Exhibit 16: Estimated New Cases of Leukemia in the US: 2020 III-7Market Analytics III-8Table 34: USA Current & Future Analysis for Cell Harvesting byType - Manual and Automated - Independent Analysis of AnnualSales in US$ Thousand for the Years 2020 through 2027 III-8

Table 35: USA Historic Review for Cell Harvesting by Type -Manual and Automated Markets - Independent Analysis of AnnualSales in US$ Thousand for Years 2012 through 2019 III-9

Table 36: USA 15-Year Perspective for Cell Harvesting by Type -Percentage Breakdown of Value Sales for Manual and Automatedfor the Years 2012, 2020 & 2027 III-10

Table 37: USA Current & Future Analysis for Cell Harvesting byApplication - Peripheral Blood, Bone Marrow, Umbilical Cord,Adipose Tissue and Other Applications - Independent Analysis ofAnnual Sales in US$ Thousand for the Years 2020 through 2027 III-11

Table 38: USA Historic Review for Cell Harvesting byApplication - Peripheral Blood, Bone Marrow, Umbilical Cord,Adipose Tissue and Other Applications Markets - IndependentAnalysis of Annual Sales in US$ Thousand for Years 2012 through2019 III-12

Table 39: USA 15-Year Perspective for Cell Harvesting byApplication - Percentage Breakdown of Value Sales forPeripheral Blood, Bone Marrow, Umbilical Cord, Adipose Tissueand Other Applications for the Years 2012, 2020 & 2027 III-13

Table 40: USA Current & Future Analysis for Cell Harvesting byEnd-Use - Biotech & Biopharma Companies, Research Institutesand Other End-Uses - Independent Analysis of Annual Sales inUS$ Thousand for the Years 2020 through 2027 III-14

Table 41: USA Historic Review for Cell Harvesting by End-Use -Biotech & Biopharma Companies, Research Institutes and OtherEnd-Uses Markets - Independent Analysis of Annual Sales in US$Thousand for Years 2012 through 2019 III-15

Table 42: USA 15-Year Perspective for Cell Harvesting byEnd-Use - Percentage Breakdown of Value Sales for Biotech &Biopharma Companies, Research Institutes and Other End-Uses forthe Years 2012, 2020 & 2027 III-16

CANADA III-17Market Overview III-17Exhibit 17: Number of New Cancer Cases in Canada: 2019 III-17Market Analytics III-18Table 43: Canada Current & Future Analysis for Cell Harvestingby Type - Manual and Automated - Independent Analysis of AnnualSales in US$ Thousand for the Years 2020 through 2027 III-18

Table 44: Canada Historic Review for Cell Harvesting by Type -Manual and Automated Markets - Independent Analysis of AnnualSales in US$ Thousand for Years 2012 through 2019 III-19

Table 45: Canada 15-Year Perspective for Cell Harvesting byType - Percentage Breakdown of Value Sales for Manual andAutomated for the Years 2012, 2020 & 2027 III-20

Table 46: Canada Current & Future Analysis for Cell Harvestingby Application - Peripheral Blood, Bone Marrow, Umbilical Cord,Adipose Tissue and Other Applications - Independent Analysis ofAnnual Sales in US$ Thousand for the Years 2020 through 2027 III-21

Table 47: Canada Historic Review for Cell Harvesting byApplication - Peripheral Blood, Bone Marrow, Umbilical Cord,Adipose Tissue and Other Applications Markets - IndependentAnalysis of Annual Sales in US$ Thousand for Years 2012 through2019 III-22

Table 48: Canada 15-Year Perspective for Cell Harvesting byApplication - Percentage Breakdown of Value Sales forPeripheral Blood, Bone Marrow, Umbilical Cord, Adipose Tissueand Other Applications for the Years 2012, 2020 & 2027 III-23

Table 49: Canada Current & Future Analysis for Cell Harvestingby End-Use - Biotech & Biopharma Companies, Research Institutesand Other End-Uses - Independent Analysis of Annual Sales inUS$ Thousand for the Years 2020 through 2027 III-24

Table 50: Canada Historic Review for Cell Harvesting by End-Use- Biotech & Biopharma Companies, Research Institutes and OtherEnd-Uses Markets - Independent Analysis of Annual Sales in US$Thousand for Years 2012 through 2019 III-25

Table 51: Canada 15-Year Perspective for Cell Harvesting byEnd-Use - Percentage Breakdown of Value Sales for Biotech &Biopharma Companies, Research Institutes and Other End-Uses forthe Years 2012, 2020 & 2027 III-26

JAPAN III-27Increasing Demand for Regenerative Medicine in GeriatricHealthcare and Cancer Care to Drive Demand for Cell Harvesting III-27Exhibit 18: Japanese Population by Age Group (2015 & 2040):Percentage Share Breakdown of Population for 0-14, 15-64 and65 & Above Age Groups III-27Exhibit 19: Cancer Related Incidence and Deaths by Site inJapan: 2018 III-28Market Analytics III-29Table 52: Japan Current & Future Analysis for Cell Harvestingby Type - Manual and Automated - Independent Analysis of AnnualSales in US$ Thousand for the Years 2020 through 2027 III-29

Table 53: Japan Historic Review for Cell Harvesting by Type -Manual and Automated Markets - Independent Analysis of AnnualSales in US$ Thousand for Years 2012 through 2019 III-30

Table 54: Japan 15-Year Perspective for Cell Harvesting by Type- Percentage Breakdown of Value Sales for Manual and Automatedfor the Years 2012, 2020 & 2027 III-31

Table 55: Japan Current & Future Analysis for Cell Harvestingby Application - Peripheral Blood, Bone Marrow, Umbilical Cord,Adipose Tissue and Other Applications - Independent Analysis ofAnnual Sales in US$ Thousand for the Years 2020 through 2027 III-32

Table 56: Japan Historic Review for Cell Harvesting byApplication - Peripheral Blood, Bone Marrow, Umbilical Cord,Adipose Tissue and Other Applications Markets - IndependentAnalysis of Annual Sales in US$ Thousand for Years 2012 through2019 III-33

Table 57: Japan 15-Year Perspective for Cell Harvesting byApplication - Percentage Breakdown of Value Sales forPeripheral Blood, Bone Marrow, Umbilical Cord, Adipose Tissueand Other Applications for the Years 2012, 2020 & 2027 III-34

Table 58: Japan Current & Future Analysis for Cell Harvestingby End-Use - Biotech & Biopharma Companies, Research Institutesand Other End-Uses - Independent Analysis of Annual Sales inUS$ Thousand for the Years 2020 through 2027 III-35

Table 59: Japan Historic Review for Cell Harvesting by End-Use -Biotech & Biopharma Companies, Research Institutes and OtherEnd-Uses Markets - Independent Analysis of Annual Sales in US$Thousand for Years 2012 through 2019 III-36

Table 60: Japan 15-Year Perspective for Cell Harvesting byEnd-Use - Percentage Breakdown of Value Sales for Biotech &Biopharma Companies, Research Institutes and Other End-Uses forthe Years 2012, 2020 & 2027 III-37

CHINA III-38Rising Incidence of Cancer Drives Cell Harvesting Market III-38Exhibit 20: Number of New Cancer Cases Diagnosed (in Thousands)in China: 2018 III-38Market Analytics III-39Table 61: China Current & Future Analysis for Cell Harvestingby Type - Manual and Automated - Independent Analysis of AnnualSales in US$ Thousand for the Years 2020 through 2027 III-39

Table 62: China Historic Review for Cell Harvesting by Type -Manual and Automated Markets - Independent Analysis of AnnualSales in US$ Thousand for Years 2012 through 2019 III-40

Table 63: China 15-Year Perspective for Cell Harvesting by Type -Percentage Breakdown of Value Sales for Manual and Automatedfor the Years 2012, 2020 & 2027 III-41

Table 64: China Current & Future Analysis for Cell Harvestingby Application - Peripheral Blood, Bone Marrow, Umbilical Cord,Adipose Tissue and Other Applications - Independent Analysis ofAnnual Sales in US$ Thousand for the Years 2020 through 2027 III-42

Table 65: China Historic Review for Cell Harvesting byApplication - Peripheral Blood, Bone Marrow, Umbilical Cord,Adipose Tissue and Other Applications Markets - IndependentAnalysis of Annual Sales in US$ Thousand for Years 2012 through2019 III-43

Table 66: China 15-Year Perspective for Cell Harvesting byApplication - Percentage Breakdown of Value Sales forPeripheral Blood, Bone Marrow, Umbilical Cord, Adipose Tissueand Other Applications for the Years 2012, 2020 & 2027 III-44

Table 67: China Current & Future Analysis for Cell Harvestingby End-Use - Biotech & Biopharma Companies, Research Institutesand Other End-Uses - Independent Analysis of Annual Sales inUS$ Thousand for the Years 2020 through 2027 III-45

Table 68: China Historic Review for Cell Harvesting by End-Use -Biotech & Biopharma Companies, Research Institutes and OtherEnd-Uses Markets - Independent Analysis of Annual Sales in US$Thousand for Years 2012 through 2019 III-46

Table 69: China 15-Year Perspective for Cell Harvesting byEnd-Use - Percentage Breakdown of Value Sales for Biotech &Biopharma Companies, Research Institutes and Other End-Uses forthe Years 2012, 2020 & 2027 III-47

EUROPE III-48Cancer in Europe: Key Statistics III-48Exhibit 21: Cancer Incidence in Europe: Number of New CancerCases (in Thousands) by Site for 2018 III-48Ageing Population to Drive Demand for Cell Harvesting Market III-49Exhibit 22: European Population by Age Group (2016, 2030 &2050): Percentage Share Breakdown by Age Group for 0-14, 15-64, and 65 & Above III-49Market Analytics III-50Table 70: Europe Current & Future Analysis for Cell Harvestingby Geographic Region - France, Germany, Italy, UK and Rest ofEurope Markets - Independent Analysis of Annual Sales in US$Thousand for Years 2020 through 2027 III-50

Table 71: Europe Historic Review for Cell Harvesting byGeographic Region - France, Germany, Italy, UK and Rest ofEurope Markets - Independent Analysis of Annual Sales in US$Thousand for Years 2012 through 2019 III-51

Table 72: Europe 15-Year Perspective for Cell Harvesting byGeographic Region - Percentage Breakdown of Value Sales forFrance, Germany, Italy, UK and Rest of Europe Markets for Years2012, 2020 & 2027 III-52

Table 73: Europe Current & Future Analysis for Cell Harvestingby Type - Manual and Automated - Independent Analysis of AnnualSales in US$ Thousand for the Years 2020 through 2027 III-53

Table 74: Europe Historic Review for Cell Harvesting by Type -Manual and Automated Markets - Independent Analysis of AnnualSales in US$ Thousand for Years 2012 through 2019 III-54

Table 75: Europe 15-Year Perspective for Cell Harvesting byType - Percentage Breakdown of Value Sales for Manual andAutomated for the Years 2012, 2020 & 2027 III-55

Table 76: Europe Current & Future Analysis for Cell Harvestingby Application - Peripheral Blood, Bone Marrow, Umbilical Cord,Adipose Tissue and Other Applications - Independent Analysis ofAnnual Sales in US$ Thousand for the Years 2020 through 2027 III-56

Table 77: Europe Historic Review for Cell Harvesting byApplication - Peripheral Blood, Bone Marrow, Umbilical Cord,Adipose Tissue and Other Applications Markets - IndependentAnalysis of Annual Sales in US$ Thousand for Years 2012 through2019 III-57

Table 78: Europe 15-Year Perspective for Cell Harvesting byApplication - Percentage Breakdown of Value Sales forPeripheral Blood, Bone Marrow, Umbilical Cord, Adipose Tissueand Other Applications for the Years 2012, 2020 & 2027 III-58

Table 79: Europe Current & Future Analysis for Cell Harvestingby End-Use - Biotech & Biopharma Companies, Research Institutesand Other End-Uses - Independent Analysis of Annual Sales inUS$ Thousand for the Years 2020 through 2027 III-59

Table 80: Europe Historic Review for Cell Harvesting by End-Use -Biotech & Biopharma Companies, Research Institutes and OtherEnd-Uses Markets - Independent Analysis of Annual Sales in US$Thousand for Years 2012 through 2019 III-60

Table 81: Europe 15-Year Perspective for Cell Harvesting byEnd-Use - Percentage Breakdown of Value Sales for Biotech &Biopharma Companies, Research Institutes and Other End-Uses forthe Years 2012, 2020 & 2027 III-61

FRANCE III-62Table 82: France Current & Future Analysis for Cell Harvestingby Type - Manual and Automated - Independent Analysis of AnnualSales in US$ Thousand for the Years 2020 through 2027 III-62

Table 83: France Historic Review for Cell Harvesting by Type -Manual and Automated Markets - Independent Analysis of AnnualSales in US$ Thousand for Years 2012 through 2019 III-63

Table 84: France 15-Year Perspective for Cell Harvesting byType - Percentage Breakdown of Value Sales for Manual andAutomated for the Years 2012, 2020 & 2027 III-64

Table 85: France Current & Future Analysis for Cell Harvestingby Application - Peripheral Blood, Bone Marrow, Umbilical Cord,Adipose Tissue and Other Applications - Independent Analysis ofAnnual Sales in US$ Thousand for the Years 2020 through 2027 III-65

Table 86: France Historic Review for Cell Harvesting byApplication - Peripheral Blood, Bone Marrow, Umbilical Cord,Adipose Tissue and Other Applications Markets - IndependentAnalysis of Annual Sales in US$ Thousand for Years 2012 through2019 III-66

Table 87: France 15-Year Perspective for Cell Harvesting byApplication - Percentage Breakdown of Value Sales forPeripheral Blood, Bone Marrow, Umbilical Cord, Adipose Tissueand Other Applications for the Years 2012, 2020 & 2027 III-67

The rest is here:
Global Cell Harvesting Market to Reach US$381,4 Million by the Year 2027 - Salamanca Press

Posted in New York Stem Cells | Comments Off on Global Cell Harvesting Market to Reach US$381,4 Million by the Year 2027 – Salamanca Press

The Canine Stem Cell Therapy Market To Go Strong With 4.2% CAGR Between 2026 – Crypto Daily

Posted: September 29, 2020 at 6:59 am

New York City, United States The change during the COVID-19 pandemic has overhauled our dependence on pattern setting developments, for instance, expanded reality, computer generated reality, and the Healthcare web of things. The unfulfilled cash related targets are persuading the relationship to grasp robotization and forefront advancements to stay ahead in the market competition. Associations are utilizing this open entryway by recognizing step by step operational needs and showing robotization in it to make an automated structure as far as might be feasible.

Persistence Market Research (PMR) has published a new research report on canine stem cell therapy. The report has been titled, Canine Stem Cell Therapy Market: Global Industry Analysis 2016 and Forecast 20172026.Veterinary research has been used in regenerative and adult stem cell therapy andhas gained significant traction over the last decade.

Canine stem cell therapy products are identified to have gained prominence over the past five years, and according to the aforementioned research report, the market for canine stem cell therapy will expand at a moderate pace over the next few years.

Get Sample Copy of Report @ https://www.persistencemarketresearch.com/samples/15550

Company Profiles

Get To Know Methodology of Report @ https://www.persistencemarketresearch.com/methodology/15550

Though all animal stem cells are not approved by FDA, veterinary stem-cell manufacturers and university researchers have been adopting various strategies in order to meet regulatory approvals, and streamline and expedite the review-and-approval process. The vendors in the market are incessantly concentrating on research and development to come up with advanced therapy, in addition to acquiring patents.

In September 2017, VetStem Biopharma, Inc. received European patent granted to the University of Pittsburgh and VetStem received full license of the patent then. This patent will eventually provide the coverage for the ongoing commercial and product development programs of VetStem and might be also available for licensing to other companies who are rather interested in this field.

The other companies operating in the global market for canine stem cell therapy are VETherapy Corporation, Aratana Therapeutics, Inc., Regeneus Ltd, Magellan Stem Cells, Animal Cell Therapies, Inc., and Medrego, among others.

According to the Persistence Market Research report, the globalcanine stem cell therapy marketis expected to witness a CAGR of 4.2% during the forecast period 2017-2026. In 2017, the market was valued at US$ 151.4 Mn and is expected to rise to a valuation of US$ 218.2 Mn by the end of 2026.

Burgeoning Prevalence of Chronic Diseases in Dogs to Benefit Market

Adipose Stem Cells (ASCs) are the most prevalent and in-demand adult stem cells owing to their safety profile, ease of harvest, and use and the ability to distinguish into multiple cell lineages. Most early clinical research is focused on adipose stem cells to treat various chronic diseases such as arthritis, tendonitis, lameness, and atopic dermatitis in dogs.

A large area of focus in veterinary medicine is treatment of osteoarthritis in dogs, which becomes more prevalent with age. Globally, more than 20% dogs are suffering from arthritis, which is a common form of canine joint and musculoskeletal disease. Out of those 20%, merely 5% seem to receive the treatment.

However, elbow dysplasia in canine registered a prevalence rate of 64%, converting it into an alarming disease condition to be treated on priority. Thereby, with the growing chronic disorders in canine, the demand for stem cell therapy is increasing at a significant pace.

Access Full Report @ https://www.persistencemarketresearch.com/checkout/15550

Expensive Nature of Therapy to Obstruct Growth Trajectory

Expensive nature and limited access to canine stem cell therapy has demonstrated to be a chief hindrance forestalling its widespread adoption. The average tier II and tier III veterinary hospitals lack the facilities and expertise to perform stem cell procedures, which necessitates the referral to a specialty vet hospital with expertise veterinarians.

A trained veterinary physician charges high treatment cost associated with stem cell therapy for dogs. Generally, dog owners have pet insurance that typically covers maximum cost associated with steam cell therapy to treat the initial injury but for the succeeding measures in case of retreatment, the costs are not covered under the pet insurance. The stem cell therapy is thus cost-prohibitive for a large number of pet owners, which highlights a major restraint to the market growth. Stem cell therapy is still in its developmental stage and a positive growth outcome for the market cannot be confirmed yet.

About- Persistence Market Research (Healthcare)

Unprecedented access to a world of information has given rise to the empowered, albeit distrustful, consumer. So is the case with healthcare, where the patient has been rendered more informed and conscientious thanks to the extraordinary penetration of the Internet. The discerning patient now demands more affordable, sophisticated, transparent, and personalized healthcare services, creating the need for new models for care.

To support companies in overcoming complex business challenges, we follow a multi-disciplinary approach. At PMR, we unite various data streams from multi-dimensional sources. By deploying real-time data collection, big data, and customer experience analytics, we deliver business intelligence for organizations of all sizes

Follow this link:
The Canine Stem Cell Therapy Market To Go Strong With 4.2% CAGR Between 2026 - Crypto Daily

Posted in Stem Cell Therapy | Comments Off on The Canine Stem Cell Therapy Market To Go Strong With 4.2% CAGR Between 2026 – Crypto Daily

Stem Cell & Regenerative Therapies | Oregon Regenerative …

Posted: July 4, 2020 at 5:46 am

Regenerative Orthopedic Medicine

Regenerative Orthopedic Medicine uses non-surgical injection procedures for the permanent repair of damaged tendons, ligaments and joints. Our regenerative and biological treatments includeProlotherapy,Platelet Rich Plasma PRP, allogenic Umbilical and Placental tissue products, andAdipose Biocellular Therapies. These treatments enhance the natural cycles of repair in chronically injured joints, ligaments, and tendons. Regenerative injections are an effective treatment for all manner of acute and chronic pain from back and neck injuries, as well as osteoarthritis of the hip, knee, shoulder, elbow, wrist, foot and ankle. Most of our patients have been told that their only solution is surgery, steroid injections, or a lifetime on pain medications. While no treatment has 100% success, the vast majority of our patients achieve durable pain-free function without surgery, joint replacement, or drugs.

Healthy patients have healthy outcomes, and that is why we practice an integrated system of Orthopedic Medicine enhanced by nutritional guidance, endocrine support, detoxification, IV Therapy, PRP, and adult stem cells in our regenerative protocols. This whole-person approach addresses the painful condition from multiple angles while promoting optimal health during the treatment process. Patients can remain active during the treatment process, and recovery time is swift.

Oregon Regenerative Medicine has found Adipose-Derived Biocellular Therapy and Platelet Rich Plasma to be effective in the following diseases:

At Oregon Regenerative Medicine, we use Platelet Rich Plasma and adipose tissues and cells that are harvested from your own body. Unlike embryonic stem cells, adipose cells and tissues have been well researched in the treatment of a wide variety of conditions. Adult-adipose tissue is the most abundant source of stem cells in the human body, and have shown great promise in the treatment of a host of conditions.

Therapies include:

Aesthetic medicine is a specialized area of medicine that focuses on improving cosmetic appearance. We focus on treatments that encourage natural collagen production. We recognize that our patients like to not only feel good but to look their best too. Our approach is to not make you look like something has been done, but rather to make you look more refreshed and younger. With regenerative medicine, you too can age gracefully!

Therapies include:

We believe that healthy patients have healthy outcomes. The foundation of functional medicine is to address the underlying cause of disease rather than simply treating symptoms. Our functional medicine service takes into account your history, lab results, medical imaging, hormone imbalance, and lifestyle to determine your optimal path to wellness.

Originally posted here:
Stem Cell & Regenerative Therapies | Oregon Regenerative ...

Posted in Oregon Stem Cells | Comments Off on Stem Cell & Regenerative Therapies | Oregon Regenerative …

Texas Stem Cell Law Opens Door for Controversial Treatments

Posted: June 11, 2020 at 4:44 am

Hospital Galenia in Cancun, Mexico, where Celltex administers autologous mesenchymal stem cells to clientsKERRY GRENSOn a main thoroughfare running along the east side of Cancun, Mexico, sits Hospital Galenia, a small, private facility with crisp, white walls and slick marble floors. On a Friday morning in February, the lobby is quiet, its palm-filled courtyard unoccupied, belying activity in parts unseen, including an emergency room and a maternity ward.

Cancuns beaches draw in visitors by the millions each year, while Galenia attracts a distinctive kind of tourist: those seeking health treatments not sanctioned by the U.S. and governments elsewhere. A number of medical tourism companies operate out of Galenia, including Houston-based Celltex Therapeutics, a company that offers stem cell therapies to mostly American customers. Celltex claims to use patients own mesenchymal stem cells (MSCs) to treat diseases as wide-ranging as amyotrophic lateral sclerosis (ALS), renal failure, and chronic obstructive pulmonary disease (COPD).

Originally, Celltex administered cells to its clients out of a laboratory site in Sugar Land, Texas. But the company had to cease offering such treatments in Texas in 2013 after the US Food and Drug Administration (FDA) sent Celltex a warning letter in 2012 stating that the patient-derived cells it was harvesting and injecting were considered drugs under US federal law, thereby requiring clinical trials and regulatory approval. The FDA review also revealed that the company mishandled the cells, failing to keep them in sterile conditions and mislabeling containers.

Rather than cede to federal laws and go the standard drug-approval route through the FDA, Celltex instead shifted its clinical operations south of the border, shipping ready-to-use MSCs in syringes to Hospital Galenia. But with a newly passed law in Texas known as Charlies Law, Celltex might have the opportunity to resume treating patients stateside. The new law could allow Texas clinics to administer certain non-FDA-approved stem cell treatments, like those offered by Celltex, to select patients.

State law is subordinate to federal FDA rules, according to the agency. Therefore, the new Texas stem cell law potentially sets up a state-federal conflict. Paul Knoepfler, a stem cell biologist at the University of California, Davis, and coauthor of a recent study that analyzed stem cell clinics in the U.S., tells The Scientist that proponents of direct-to-consumer stem cell clinics could use the law as an opportunity to challenge the FDAs authority, which could result in a federal court case.

While Texas is loosening regulations on unproven stem cell treatments, the FDA and health authorities in other countries are going in the opposite direction.

Celltexwhose board of directors includes Texass former first lady, Anita Thigpen Perrywas actively involved in the bill, according to a Celltex spokesperson. The law is a step in the right direction, according to the firm, but still far from a law which would allow us to provide high-dose adult stem cell banking and therapy in the United States.

Direct-to-consumer stem cell clinics, influential Texas politicianssome with ties to Celltexand some patients eager for alternative therapeutic options applaud the law. But scientists question the cells efficacy, and bioethicists voice concerns about selling a therapy before the evidence is all in. Without rigorous testing and FDA oversight, critics say, there are no guarantees that these direct-to-consumer stem cell clinics abide by rigorous laboratory practices and that the interventions are not putting patients at risk.

Its true that we are very excited about the ability to develop effective new therapies using stem cells but there is a tremendous amount of research required to figure out how to do this effectively and safely, says Sean Morrison, a stem cell biologist and director of the Childrens Medical Center Research Institute at the University of Texas Southwestern Medical Center in Dallas. There are many companies short-circuiting that process and selling patients unproven entities that they have not shown to be effective nor safe. In some cases, the unproven therapies are scientifically implausible based on what we know about the biology.

According to Celltex, the company treats about 400 to 500 people each year using MSCs harvested from the patients own belly fat. The cells are then purified, expanded, and frozen in Texas before being infused intravenously or injected into the affected tissue. According to the medical director of Celltexs Galenia clinic, Gabriel Salazar, most patients come for relief of inflammatory conditions, especially arthritis. There are people who come for wellness as well, says Salazar. They arent sick, but they have risk factors.

The physiological mechanism of MSCs is unclear. Salazar says they initiate anti-inflammatory pathways, a phenomenon that researchers have observed in vitro in the lab. From animal models, scientists understand that these are cells that regulate the microenvironment at sites of injury . . . but no one really understands how they function in vivo, says Arnold Caplan, an MSC researcher at Case Western University.

All [of our clients] have received improvements, some minimally and some miraculously. And no one has had a problem.David Eller,CEO, Celltex

Clinicaltrials.gov lists 243 ongoing MSC trials, including some in the U.S., sponsored by industry, the National Institutes of Health, and others. Yet there are no FDA-approved therapies using MSCs for any condition. The FDA, for its part, is studying MSCs to develop standardized isolation methods and to learn how MSCs behave when injected into the body.

Anecdotal testimonials from clients of Celltex and other firms abound, but rigorous clinical evidence is hard to come by, as companies selling stem cell therapies dont often publish in peer-reviewed journals. In fact, there are no published data on MSCs efficacy in ameliorating arthritis or the numerous other conditions that patients pay Celltex to treat. Thus far, Celltex has one publication documenting MSC use in patientsa 2017 case report of two individuals with a nervous system disorder that suggests the autologous adipose-derived MSCs they received resulted in symptom improvements.

Celltex CEO David Eller says the company has safely administered MSCs to more than 4,500 clients. All of them have received improvements, some minimally and some miraculously, he tells The Scientist. And no one has had a problem. In opposition to the FDAs stance, Eller considers clinical trials a waste of patients time. We can wait another decade or more for costly clinical trials and watch as our loved ones deteriorate, or we can do something about it now. In our eyes, doing nothing is the real risk, he writes on the Celltex website.

Nevertheless, Celltex has pledged to conduct its first clinical trial. Just last month (February 27), the firm announced that the Comisin Federal Para La Proteccin Contra Riesgos Sanitarios Comisin de Autorizacin Sanitaria (COFEPRIS, the Mexican equivalent of the US FDA) had given the go-ahead for Galenia to start a clinical trial in patients with osteoarthritis and rheumatoid arthritis. The Phase 2 study will use Celltexs protocols to generate cells in quantities never possible for use in therapies for vascular, autoimmune, and degenerative diseases, as well as injuries, according to a company press release.

Details on the trial are scant. The press release states that the trial will measure the quality of life (QoL) of subjects prior to and after receiving the therapy. Celltex tells The Scientist that the trial will have a safety and toxicity monitoring component and that participants will not incur cost of the cell procedure. According to the Celltex website, the company charges $6,500 to obtain, process, and store a clients MSCs for one year, while the pricing for an injection varies depending on each individuals situation.

Im all for investigating QoL self-assessments in the context of clinical research, says Leigh Turner, a bioethicist at the University of Minnesota who studies direct-to-consumer stem cell clinics, but one obvious concern in the case of this particular study is that it will do little more than capture a placebo [effect].

According to Turner, Celltex is not the first US business selling unapproved stem cell interventions to conduct a QoL study rather than one specifically designed to test for safety and efficacy endpoints. For marketing approval, the FDA requires that stem cell products be tested for safety and efficacy and that the company developing the product file an IND application with the agency. Yet trials can be listed on registries such as clinicaltrials.gov whether or not they comply with FDA regulatory standards and safety oversight.

I hope [Celltex] pursues an Investigational New Drug application [IND] in the U.S. and conducts a placebo-controlled, double-blinded study, writes Knoepfler in an email to The Scientist. If their product is safe and clearly effective, such a rigorous study could prove that fairly conclusively.

Human mesenchymal stem cells with nuclei labeled blue and actin filaments greenFLICKR, ENGINEERING AT CAMBRIDGE

The new Texas stem cell law has yet to go into effect; the states Health and Human Services Commission is still developing the procedures of how the law will be implemented. As Celltex and other direct-to-consumer stem cell companies wait for lawmakers to clarify details on the new rules, Celltex will continue with its cell-banking operations in Texas and its infusions in Mexico, a Celltex spokesperson tells The Scientist.

According to State Senator Paul Bettencourt (R-Houston), an author of the bill, the plan is for each stem cell treatment course to be reviewed by an Independent Review Board (IRB). After a favorable review, the treatment could then be administered in one of the hundreds of medical institutions in Texas.

The Texas law is unique in its permissiveness, says Turner. A new law in California, which took effect January 1, requires that direct-to-consumer stem cell clinics disclose to potential customers that their products are not approved by the FDA. Other states appear to be looking for ways to better protect patients and consumers instead of looking for ways to lower regulatory standards, writes Turner.

While Texas is loosening regulations on unproven stem cell treatments, the FDA and health authorities in other countries are going in the opposite direction, focusing more oversight and scrutiny on direct-to-consumer stem cell clinics. The FDA has recently warned consumers about stem cell clinics touting unproven treatments, and in August 2017, the FDA commissioner announced plans for better enforcement of federal laws and oversight of stem cell clinics. Australia recently announced a higher level of stem cell clinic regulation, as have health agencies in Canada and India.

To be a candidate to receive the therapy under Charlies law, the patient must have a severe, chronic disease or be terminally ill. And the treatment must be registered in a clinical trial database somewhere in the world. According to Bettencourt, the next step is for the Texas Medical Board to draw up the process for how to evaluate and oversee each patients case.

But the bill does not include any details on product oversight or quality control, and reporting by The Scientist reveals questionable practices.

When The Scientist spoke to Eller, he said that Celltex uses the MD Anderson Flow Cytometry Facility weekly to validate that the cells are 100 percent patient-derived MSCs. And in follow-up emails, a spokesperson for Celltex also stated that members of the MD Anderson core staff independently conduct quality control and validation.

However, the cytometry facility had a different story.

Scott Melville, an MD Anderson spokesperson, tells The Scientist that Celltex has paid to use the public Flow Cytometry Facility since 2015, but that that no one at the facility independently assesses the quality and content of the [Celltex] samples. According to MD Andersons recent records from the last several months up to March 23, 2018, Celltex used the facility for two hours on November 2, 2017, and for one hour each on January 18 and January 19, 2018.

Critics of the Texas stem cell law say that the language in the bill is too vague and are skeptical that there will be enough independent scrutiny to make sure patients are not exposed to unsafe treatments. Im concerned that businesses could use it to make money by putting patients at risk, says Knoepfler.

Reporting from Mexico by Kerry Grens

Read more:
Texas Stem Cell Law Opens Door for Controversial Treatments

Posted in Texas Stem Cells | Comments Off on Texas Stem Cell Law Opens Door for Controversial Treatments

Covid 19 Outbreak Cell Harvesting System Market 2020 Product Type, Applications/end user, Key Players and Geographical Regions 2026 – Jewish Life…

Posted: June 4, 2020 at 9:10 am

COVID-19 impact will also be included and considered for forecast.

Global Cell Harvesting System Market research report provides detail information about Market Introduction, Market Summary, Global market Revenue (Revenue USD), Market Drivers, Market Restraints, Market Opportunities, Competitive Analysis, Regional and Country Level.

Cell Harvesting System Market Size Covers Global Industry Analysis, Size, Share, CAGR, Trends, Forecast And Business Opportunity.

>>Need a PDF of the global market report? Visit: https://industrystatsreport.com/Request/Sample?ResearchPostId=153&RequestType=Sample

Cell Harvesting System Market: Increase in healthcare facilities and increase in bone marrow transplantation are key drivers for the Global Cell Harvesting System Market.

The global cell harvesting systems market size was valued USD 3533.27 Million in 2017 and is expected to grow at a CAGR of 14.01% over the forecast period.

Cell harvesting is a system which is used to cultivate, regenerate, transplant and repair the damages organs with the healthy one. Cell harvesting is one of the important parts of biopharmaceutical industry which directly relates with the quality of product. Stem cell harvesting also helps in the treatment of various diseases such as cancer, autoimmune disease, anemia and others. So, during the study of Global Cell Harvesting System market, we have considered Cell Harvesting System to analyze the market.

Global Cell Harvesting System Market report is segmented on the technique type, application type, end user type and by regional & country level. Based upon technique type, global Cell Harvesting System Market is classified as Altered Nuclear Transfer and Blastomere Extraction. Based upon Application type, global Cell Harvesting System Market is classified as Bone Marrow, Peripheral Blood, Umbilical Cord Blood, and Adipose Tissue. Based upon end users, global Cell Harvesting System Market is classified as Research Centers, Academics Institutes, Diagnostic Labs, and Hospitals.

The regions covered in this Cell Harvesting System Market report are North America, Europe, Asia-Pacific and Rest of the World. On the basis of country level, market of Cell Harvesting System is sub divided into U.S., Mexico, Canada, U.K., France, Germany, Italy, China, Japan, India, South East Asia, GCC, Africa, etc.

Key Players for Global Cell Harvesting System Market Reports

Global Cell Harvesting System market report covers prominent players like Tomtec, Bertin Technologies, PerkinElmer Inc., TERUMO BCT, INC., SP Scienceware, hynoDent AG, Avita Medical, BRAND GMBH Teleflex Incorporated., Argos Technologies, Inc., Thomas Scientific, Arthrex, Inc. and others.

Global Cell Harvesting System Market Dynamics

The commercialization and growth of global Cell Harvesting System market over the past 25 years has been highly impactful. Bone marrow transplantation is one of the major factors driving the growth of cell harvesting system over the forecast period. Due to the increase in blood cancer it has raised the demand for bone marrow transplantation which in turn increased the demand for cell harvesting system. As per The Leukemia & Lymphoma Society report 2018, an estimated combined total of 174,250 people in the US are expected to be diagnosed with leukemia, lymphoma or myeloma in 2018. There is also an increase in awareness about stem cells and its advantages which are helpful in the treatment of various disorders. Furthermore, various technological advancement have also increase the new and better technologies with better results are expected to promote the growth of cell harvesting system market over the forecast period. However, High cost, lack of reimbursement policies, immune rejection and others are the various factors which are expected to hamper the growth of cell harvesting system market over the forecast period.

Global Cell Harvesting System Market Regional Analysis

North America dominates the market with highest market share which is closely followed by the Europe over the forecast period. Due to the increased prevalence of leukemia, lymphoma and others coupled with increased healthcare facilities. As per The Leukemia & Lymphoma Society 2018 report, new cases of leukemia, lymphoma and myeloma are expected to account for 10 percent of the estimated 1,735,350 new cancer cases diagnosed in the US in 2018. Asia Pacific is expected to be the third largest and fastest growing region over the forecast period. Due to various technological advancements, increase in awareness among people and others are expected to support the growth of cell harvesting system market over the forecast period. Furthermore, Increase in healthcare facilities in the developing economies such as India, China and others are expected to fuel the growth of cell harvesting system market. Latin America, Middle East and Africa and expected to develop at a considerable rate over the forecast period.

Key Benefits for Global Cell Harvesting System Market Reports

Global Cell Harvesting System market report covers in depth historical and forecast analysis.Global Cell Harvesting System Market research report provides detail information about Market Introduction, Market Summary, Global market Revenue (Revenue USD), Market Drivers, Market Restraints, Market opportunities, Competitive Analysis, Regional and Country Level.Global Cell Harvesting System Market report helps to identify opportunities in market place.Global Cell Harvesting System Market report covers extensive analysis of emerging trends and competitive landscape.

By Techniques Type:

Altered Nuclear TransferBlastomere Extraction

By Application:

Bone MarrowPeripheral BloodUmbilical Cord BloodAdipose Tissue

By End User:

Research CentersAcademics InstitutesDiagnostic LabsHospitals

By Region

North AmericaU.S.CanadaEuropeUKFranceGermanyItalyAsia PacificChinaJapanIndiaSoutheast AsiaLatin AmericaBrazilMexicoThe Middle East and AfricaGCCAfricaRest of Middle East and Africa

Cell Harvesting System Market Key PlayersTomtecBertin TechnologiesPerkinElmer Inc.TERUMO BCT, INC.SP SciencewarehynoDent AGAvita MedicalBRAND GMBHTeleflex Incorporated.Argos Technologies, Inc.Thomas ScientificArthrex, Inc.

Need a PDF of the global market report? Visit: https://industrystatsreport.com/Request/Sample?ResearchPostId=153&RequestType=Methodology

Table of Content:

Market Overview: The report begins with this section where product overview and highlights of product and application segments of the Global Cell Harvesting System Market are provided. Highlights of the segmentation study include price, revenue, sales, sales growth rate, and market share by product.

Competition by Company: Here, the competition in the Worldwide Global Cell Harvesting System Market is analyzed, By price, revenue, sales, and market share by company, market rate, competitive situations Landscape, and latest trends, merger, expansion, acquisition, and market shares of top companies.

Company Profiles and Sales Data: As the name suggests, this section gives the sales data of key players of the Global Cell Harvesting System Market as well as some useful information on their business. It talks about the gross margin, price, revenue, products, and their specifications, type, applications, competitors, manufacturing base, and the main business of key players operating in the Global Cell Harvesting System Market.

Market Status and Outlook by Region: In this section, the report discusses about gross margin, sales, revenue, production, market share, CAGR, and market size by region. Here, the Global Cell Harvesting System Market is deeply analyzed on the basis of regions and countries such as North America, Europe, China, India, Japan, and the MEA.

Application or End User: This section of the research study shows how different end-user/application segments contribute to the Global Cell Harvesting System Market.

Market Forecast: Here, the report offers a complete forecast of the Global Cell Harvesting System Market by product, application, and region. It also offers global sales and revenue forecast for all years of the forecast period.

Research Findings and Conclusion: This is one of the last sections of the report where the findings of the analysts and the conclusion of the research study are provided.

About Us:

We publish market research reports & business insights produced by highly qualified and experienced industry analysts. Our research reports are available in a wide range of industry verticals including aviation, food & beverage, healthcare, ICT, Construction, Chemicals and lot more. Brand Essence Market Research report will be best fit for senior executives, business development managers, marketing managers, consultants, CEOs, CIOs, COOs, and Directors, governments, agencies, organizations and Ph.D. Students.

Top Trending Reports:

Top Trending Reports:

Read more from the original source:
Covid 19 Outbreak Cell Harvesting System Market 2020 Product Type, Applications/end user, Key Players and Geographical Regions 2026 - Jewish Life...

Posted in New Mexico Stem Cells | Comments Off on Covid 19 Outbreak Cell Harvesting System Market 2020 Product Type, Applications/end user, Key Players and Geographical Regions 2026 – Jewish Life…

Canine Stem Cell Therapy Market Covid-19 Impact In 2026 | In-depth Analysis, Global Market Share, Top Trends, Professional & Technical Industry…

Posted: May 30, 2020 at 3:56 am

New York City, United States Since the COVID-19 infection flare-up in December 2019, the malady has spread to right around 100 nations around the world with the World Health Organization proclaiming it a general wellbeing crisis. The worldwide effects of the coronavirus sickness 2019 (COVID-19) are now beginning to be felt, and will essentially influence the Healthcare Industry in 2020.

Persistence Market Research (PMR) has published a new research report on canine stem cell therapy. The report has been titled, Canine Stem Cell Therapy Market: Global Industry Analysis 2016 and Forecast 20172026.Veterinary research has been used in regenerative and adult stem cell therapy andhas gained significant traction over the last decade.

Canine stem cell therapy products are identified to have gained prominence over the past five years, and according to the aforementioned research report, the market for canine stem cell therapy will expand at a moderate pace over the next few years.

Get Sample Copy of Report @ https://www.persistencemarketresearch.com/samples/15550

Company Profiles

Get To Know Methodology of Report @ https://www.persistencemarketresearch.com/methodology/15550

Though all animal stem cells are not approved by FDA, veterinary stem-cell manufacturers and university researchers have been adopting various strategies in order to meet regulatory approvals, and streamline and expedite the review-and-approval process. The vendors in the market are incessantly concentrating on research and development to come up with advanced therapy, in addition to acquiring patents.

In September 2017, VetStem Biopharma, Inc. received European patent granted to the University of Pittsburgh and VetStem received full license of the patent then. This patent will eventually provide the coverage for the ongoing commercial and product development programs of VetStem and might be also available for licensing to other companies who are rather interested in this field.

The other companies operating in the global market for canine stem cell therapy are VETherapy Corporation, Aratana Therapeutics, Inc., Regeneus Ltd, Magellan Stem Cells, Animal Cell Therapies, Inc., and Medrego, among others.

According to the Persistence Market Research report, the globalcanine stem cell therapy marketis expected to witness a CAGR of 4.2% during the forecast period 2017-2026. In 2017, the market was valued at US$ 151.4 Mn and is expected to rise to a valuation of US$ 218.2 Mn by the end of 2026.

Burgeoning Prevalence of Chronic Diseases in Dogs to Benefit Market

Adipose Stem Cells (ASCs) are the most prevalent and in-demand adult stem cells owing to their safety profile, ease of harvest, and use and the ability to distinguish into multiple cell lineages. Most early clinical research is focused on adipose stem cells to treat various chronic diseases such as arthritis, tendonitis, lameness, and atopic dermatitis in dogs.

A large area of focus in veterinary medicine is treatment of osteoarthritis in dogs, which becomes more prevalent with age. Globally, more than 20% dogs are suffering from arthritis, which is a common form of canine joint and musculoskeletal disease. Out of those 20%, merely 5% seem to receive the treatment.

However, elbow dysplasia in canine registered a prevalence rate of 64%, converting it into an alarming disease condition to be treated on priority. Thereby, with the growing chronic disorders in canine, the demand for stem cell therapy is increasing at a significant pace.

Access Full Report @ https://www.persistencemarketresearch.com/checkout/15550

Expensive Nature of Therapy to Obstruct Growth Trajectory

Expensive nature and limited access to canine stem cell therapy has demonstrated to be a chief hindrance forestalling its widespread adoption. The average tier II and tier III veterinary hospitals lack the facilities and expertise to perform stem cell procedures, which necessitates the referral to a specialty vet hospital with expertise veterinarians.

A trained veterinary physician charges high treatment cost associated with stem cell therapy for dogs. Generally, dog owners have pet insurance that typically covers maximum cost associated with steam cell therapy to treat the initial injury but for the succeeding measures in case of retreatment, the costs are not covered under the pet insurance. The stem cell therapy is thus cost-prohibitive for a large number of pet owners, which highlights a major restraint to the market growth. Stem cell therapy is still in its developmental stage and a positive growth outcome for the market cannot be confirmed yet.

Explore Extensive Coverage of PMR`sLife Sciences & Transformational HealthLandscape

Pressure Guidewires MarketPressure Guidewire Market Segmented By Flat Tipped Pressure Guidewires, Flexible Tipped Pressure Guidewires Product with Pressure Wire Technology, Optical Fiber Technology Type for Hospitals, Ambulatory Surgical Centers, Independent Catheterization Labs.For More Information

Compression Therapy Devices MarketThe compression therapy market of North America is projected to escalate from US$ 2,377.8 Mn in 2014 to US$ 1,515.4 Mn, registering a CAGR of 5.4% by 2020-end.For More Information

About us:

Persistence Market Research (PMR) is a third-platform research firm. Our research model is a unique collaboration of data analytics andmarket research methodologyto help businesses achieve optimal performance.

To support companies in overcoming complex business challenges, we follow a multi-disciplinary approach. At PMR, we unite various data streams from multi-dimensional sources. By deploying real-time data collection, big data, and customer experience analytics, we deliver business intelligence for organizations of all sizes.

Our client success stories feature a range of clients from Fortune 500 companies to fast-growing startups. PMRs collaborative environment is committed to building industry-specific solutions by transforming data from multiple streams into a strategic asset.

Contact us:

Ashish KoltePersistence Market ResearchAddress 305 Broadway, 7th FloorNew York City,NY 10007 United StatesU.S. Ph. +1-646-568-7751USA-Canada Toll-free +1 800-961-0353Salessales@persistencemarketresearch.comWebsitehttps://www.persistencemarketresearch.com

See more here:
Canine Stem Cell Therapy Market Covid-19 Impact In 2026 | In-depth Analysis, Global Market Share, Top Trends, Professional & Technical Industry...

Posted in Stem Cell Therapy | Comments Off on Canine Stem Cell Therapy Market Covid-19 Impact In 2026 | In-depth Analysis, Global Market Share, Top Trends, Professional & Technical Industry…

Stem cell therapy: a potential approach for treatment of influenza virus and coronavirus-induced acute lung injury – BMC Blogs Network

Posted: May 29, 2020 at 9:48 am

Acute lung injury (ALI) is a devastating disease process involving pulmonary edema and atelectasis caused by capillary membrane injury [1]. The main clinical manifestation is the acute onset of hypoxic respiratory failure, which can subsequently trigger a cascade of serious complications and even death [2]. Thus, ALI causes a considerable financial burden for health care systems throughout the world. ALI can result from various causes, including multiple traumas, large-volume blood transfusions, and bacterial and viral infections [2, 3]. A variety of viruses, including influenza virus, coronavirus (CoV), adenovirus, cytomegalovirus (CMV), and respiratory syncytial virus (RSV), are associated with ALI [4]. Importantly, most viruses, whose hosts are various animal species, can cause severe and rapidly spreading human infections. In the early 2000s, several outbreaks of influenza virus and CoV emerged, causing human respiratory and intestinal diseases worldwide, including the more recent SARS-CoV-2 infection [5,6,7]. To date, SARS-CoV-2 has affected more than 80,000 people, causing nearly 3300 deaths in China and more than 1,800,000 people, causing nearly 110,000 deaths all over the world (http://2019ncov.chinacdc.cn/2019-nCoV/).

Infectious respiratory diseases caused by different viruses are associated with similar respiratory symptoms ranging from the common cold to severe acute respiratory syndrome [8]. This makes the clinical distinction between different agents involved in infection very difficult [8, 9]. Currently, the clinical experience mainly includes antibacterial and antiviral drug treatment derived from handling several outbreaks of influenza virus and human CoVs. Numerous agents have been identified to inhibit the entry and/or replication of these viruses in cell culture or animal models [10]. Although these antiviral drugs can effectively prevent and eliminate the virus, the full recovery from pneumonia and ALI depends on the resistance of the patient. Recently, stem cell-based therapy has become a potential approved tool for the treatment of virus-induced lung injury [11,12,13]. Here, we will give a brief overview of influenza virus and CoVs and then present the cell-based therapeutic options for lung injury caused by different kinds of viruses.

Influenza virus and human CoV are the two most threatening viruses for infectious lung injury [14]. These pathogens can be transmitted through direct or indirect physical contact, droplets, or aerosols, with increasing evidence suggesting that airborne transmission, including via droplets or aerosols, enhances the efficiency of viral transmission among humans and causes uncontrolled infectious disease [15]. Throughout human history, outbreaks and occasional pandemics caused by influenza virus and CoV have led to approximately hundreds of millions of deaths worldwide [16].

Influenza virus is a well-known human pathogen that has a negative-sense RNA genome [17]. According to its distinct antigenic properties, the influenza virus can be divided into 4 subtypes, types A, B, C, and D. Influenza A virus (IAV) lineages in animal populations cause economically important respiratory disease. Little is known about the other human influenza virus types B, C, and D [18]. Further subtypes are characterized by the genetic and antigenic properties of the hemagglutinin (HA) and neuraminidase (NA) glycoproteins [19]. Sporadic and seasonal infections in swine with avian influenza viruses of various subtypes have been reported. The most recent human pandemic virusesH1N1 from swine and H5N1 from aviancause severe respiratory tract disease and lung injury in humans [20, 21].

CoVs, a large family of single-stranded RNA viruses, typically affect the respiratory tract of mammals, including humans. CoVs are further divided into four genera: alpha-, beta-, gamma-, and delta-CoVs. Alpha- and beta-CoVs can infect mammals, and gamma- and delta-CoVs tend to infect birds, but some of these viruses can also be transmitted to mammals [22]. Human CoVs were considered relatively harmless respiratory pathogens in the past. Infections with the human CoV strains 229E, OC43, NL63, and HKU1 usually result in mild respiratory illness, such as the common cold [23]. In contrast, the CoV responsible for the 2002 severe acute respiratory syndrome (SARS-CoV), the 2012 Middle East respiratory syndrome CoV (MERS-CoV), and, more recently, the SARS-CoV-2 have received global attention owing to their genetic variation and rapid spread in human populations [5,6,7].

Usually, the influenza virus can enter the columnar epithelial cells of the respiratory tract, such as the trachea, bronchi, and bronchioles. Subsequently, the influenza virus begins to replicate for an asymptomatic period of time and then migrate to the lung tissue to cause acute lung and respiratory injury [24]. Similar to those with influenza virus infection, patients with SARS, MERS, or SARS-CoV-2 present with various clinical features, ranging from asymptomatic or mild respiratory illness to severe ALI, even with multiple organ failure [5,6,7]. The pathogenesis of ALI caused by influenza virus and human CoV is often associated with rapid viral replication, marked inflammatory cell infiltration, and elevated proinflammatory cytokine/chemokine responses [25]. Interestingly, in IAV- and human CoV-infected individuals, the pulmonary pathology always involves diffuse alveolar damage, but viral RNA is present in only a subset of patients [26]. Some studies suggest that an overly exaggerated immune response, rather than uncontrolled viral spread, is the primary cause in fatal cases caused by virus infection [27]. Several immune cell types have been found to contribute to damaging host responses, providing novel approaches for therapeutic intervention [28].

IAV infection, the most common cause of viral pneumonia, causes substantial seasonal and pandemic morbidity and mortality [29]. Currently, antiviral drugs are the primary treatment strategy for influenza-induced pneumonia. However, antiviral drugs cannot repair damaged lung cells. Here, we summarize the present studies of stem cell therapy for influenza virus-induced lung injury.

Mesenchymal stem/stromal cells (MSCs) constitute a heterogeneous subset of stromal regenerative cells that can be harvested from several adult tissue types, including bone marrow, umbilical cord, adipose, and endometrium [30]. They retain the expression of the markers CD29, CD73, CD90, and CD105 and have a rapid proliferation rate, low immunogenicity, and low tumorigenicity [30]. MSCs also have self-renewal and multidifferentiation capabilities and exert immunomodulatory and tissue repair effects by secreting trophic factors, cytokines, and chemokines [31]. Due to these beneficial biological properties, MSCs and their derivatives are attractive as cellular therapies for various inflammatory diseases, including virus-induced lung injury.

Several studies on IAV-infected animal models have shown the beneficial effects of the administration of different tissue-derived MSCs [32,33,34,35]. H5N1 virus infection reduces alveolar fluid clearance (AFC) and enhances alveolar protein permeability (APP) in human alveolar epithelial cells, which can be inhibited by coculture with human bone marrow-derived MSCs (BMSCs) [32]. Mechanistically, this process can be mediated by human BMSC secreted angiopoietin-1 (Ang1) and keratinocyte growth factor (KGF) [32]. Moreover, in vivo experiments have shown that human BMSCs have a significant anti-inflammatory effect by increasing the number of M2 macrophages and releasing various cytokines and chemokines, such as interleukin (IL)-1, IL-4, IL-6, IL-8, and IL-17 [32]. Similar anti-inflammatory effects have been achieved in another virus-induced lung injury model. The intravenous injection of mouse BMSCs into H9N2 virus-infected mice significantly attenuates H9N2 virus-induced pulmonary inflammation by reducing chemokine (GM-CSF, MCP-1, KC, MIP-1, and MIG) and proinflammatory cytokine (IL-1, IL-6, TNF-, and IFN-) levels, as well as reducing inflammatory cell recruitment into the lungs [33]. Another study on human BMSCs cocultured with CD8+ T cells showed that MSCs inhibit the proliferation of virus-specific CD8+ T cells and the release of IFN- by specific CD8+ T cells [36].

In addition, human umbilical cord-derived MSCs (UC-MSCs) were found to have a similar effect as BMSCs on AFC, APP, and inflammation by secreting growth factors, including Ang1 and hepatocyte growth factor (HGF), in an in vitro lung injury model induced by H5N1 virus [34]. UC-MSCs also promote lung injury mouse survival, increase the body weight, and decreased the APP levels and inflammation in vivo [34]. Unlike Ang1, KGF, and HGF mentioned above, basic fibroblast growth factor 2 (FGF2) plays an important role in lung injury therapy via immunoregulation. The administration of the recombinant FGF2 protein improves H1N1-induced mouse lung injury and promotes the survival of infected mice by recruiting and activating neutrophils via the FGFR2-PI3K-AKT-NFB signaling pathway [37]. FGF2-overexpressing MSCs have an enhanced therapeutic effect on lipopolysaccharide-induced ALI, as assessed by the proinflammatory factor level, neutrophil quantity, and histopathological index of the lung [38].

MSCs secrete various soluble factors and extracellular vesicles (EVs), which carry lipids, proteins, DNA, mRNA, microRNAs, small RNAs, and organelles. These biologically active components can be transferred to recipient cells to exert anti-inflammatory, antiapoptotic, and tissue regeneration functions [39]. EVs isolated from conditioned medium of pig BMSCs have been demonstrated to have anti-apoptosis, anti-inflammation, and antiviral replication functions in H1N1-affected lung epithelial cells and alleviate H1N1-induced lung injury in pigs [35]. Moreover, the preincubation of EVs with RNase abrogates their anti-influenza activity, suggesting that the anti-influenza activity of EVs is due to the transfer of RNAs from EVs to epithelial cells [35]. Exosomes are a subset of EVs that are 50200nm in diameter and positive for CD63 and CD81 [40]. Exosomes isolated from the conditioned medium of UC-MSCs restore the impaired AFC and decreased APP in alveolar epithelial cells affected by H5N1 virus [34]. In addition, the ability of UC-MSCs to increase AFC is superior to that of exosomes, which indicates that other components secreted by UC-MSCs have synergistic effects with exosomes [34].

Despite accumulating evidence demonstrating the therapeutic effects of MSC administration in various preclinical models of lung injury, some studies have shown contrasting results. Darwish and colleagues proved that neither the prophylactic nor therapeutic administration of murine or human BMSCs could decrease pulmonary inflammation or prevent the progression of ALI in H1N1 virus-infected mice [41]. In addition, combining MSC administration with the antiviral agent oseltamivir was also found to be ineffective [41]. Similar negative results were obtained in another preclinical study. Murine or human BMSCs were administered intravenously to H1N1-induced ARDS mice [42]. Although murine BMSCs prevented influenza-induced thrombocytosis and caused a modest reduction in lung viral load, murine or human BMSCs failed to improve influenza-mediated lung injury as assessed by weight loss, the lung water content, and bronchoalveolar lavage inflammation and histology, which is consistent with Darwishs findings [42]. However, the mild reduction in viral load observed in response to murine BMSC treatment suggests that, on balance, MSCs are mildly immunostimulatory in this model [42]. Although there are some controversial incidents in preclinical research, the transplant of menstrual-blood-derived MSCs into patients with H7N9-induced ARDS was conducted at a single center through an open-label clinical trial (http://www.chictr.org.cn/). MSC transplantation significantly lowered the mortality and did not result in harmful effects in the bodies of the patients [43]. This clinic study suggests that MSCs significantly improve the survival rate of influenza virus-induced lung injury.

The effects of exogenous MSCs are exerted through their isolation and injection into test animals. There are also some stem/progenitor cells that can be activated to proliferate when various tissues are injured. Basal cells (BCs), distributed throughout the pseudostratified epithelium from the trachea to the bronchioles, are a class of multipotent tissue-specific stem cells from various organs, including the skin, esophagus, and olfactory and airway epithelia [44, 45]. Previously, TPR63+/KRT5+ BCs were shown to self-renew and divide into club cells and ciliated cells to maintain the pseudostratified epithelium of proximal airways [46]. Several studies have shown that TPR63+/KRT5+ BCs play a key role in lung repair and regeneration after influenza virus infection. When animals typically recover from H1N1 influenza infection, TPR63+/KRT5+ BCs accumulate robustly in the lung parenchyma and initiate an injury repair process to maintain normal lung function by differentiating into mature epithelium [47]. Lineage-negative epithelial stem/progenitor (LNEP) cells, present in the normal distal lung, can activate a TPR63+/KRT5+ remodeling program through Notch signaling after H1N1 influenza infection [48]. Moreover, a population of SOX2+/SCGB1A/KRT5 progenitor cells can generate nascent KRT5+ cells as an early response to airway injury upon H1N1 influenza virus infection [49]. In addition, a rare p63+Krt5 progenitor cell population also responds to H1N1 virus-induced severe injury [50]. This evidence suggests that these endogenous lung stem/progenitor cells (LSCs) play a critical role in the repopulation of damaged lung tissue following severe influenza virus infection (Table2).

Taken together, the present in vitro (Table1) and in vivo (Table2) results show that MSCs and LSCs are potential cell sources to treat influenza virus-induced lung injury.

Lung injury caused by SARS, MERS, or SARS-CoV-2 poses major clinical management challenges because there is no specific treatment that has been proven to be effective for each infection. Currently, virus- and host-based therapies are the main methods of treatment for spreading CoV infections. Virus- and host-based therapies include monoclonal antibodies and antiviral drugs that target the key proteins and pathways that mediate viral entry and replication [51].The major challenges in the clinical development of novel drugs include a limited number of suitable animal models for SARS-CoV, MERS-CoV, and SARS-CoV-2 infections and the current absence of new SARS and MERS cases [51]. Although the number of cases of SARS-CoV-2-induced pneumonia patients is continuously increasing, antibiotic and antiviral drugs are the primary methods to treat SARS-CoV-2-infected patients. Similar to that of IAV, human CoV-mediated damage to the respiratory epithelium results from both intrinsic viral pathogenicity and a robust host immune response. The excessive immune response contributes to viral clearance and can also worsen the severity of lung injury, including the demise of lung cells [52]. However, the present treatment approaches have a limited effect on lung inflammation and regeneration.

Stem cell therapy for influenza virus-induced lung injury shows promise in preclinical models. Although it is difficult to establish preclinical models of CoV-induced lung injury, we consider stem cell therapies to be effective approaches to improve human CoV-induced lung injury. Acute inflammatory responses are one of the major underlying mechanisms for virus-induced lung injury. Innate immune cells, including neutrophils and inflammatory monocytes-macrophages (IMMs), are major innate leukocyte subsets that protect against viral lung infections [53]. Both neutrophils and IMMs are rapidly recruited to the site of infection and play crucial roles in the host defense against viruses. Neutrophils and IMMs can activate Toll-like receptors (TLRs) and produce interferons (IFNs) and other cytokines/chemokines [54]. There are two functional effects produced by the recruitment of neutrophils and IMMs: the orchestration of effective adaptive T cell responses and the secretion of inflammatory cytokines/chemokines [55]. However, excessive inflammatory cytokine and chemokine secretion impairs antiviral T cell responses, leading to ineffective viral clearance and reduced survival [56].

MSCs are known to suppress both innate and adaptive immune responses. MSCs have been suggested to inhibit many kinds of immune cells, including T cells, B cells, dendritic cells (DCs), and natural killer (NK) cells in vitro and in vivo [57] (Fig.1). Several molecules, including IL-1, TNF-, and INF-, most of which are produced by inflammatory cells, are reported to be involved in MSC-mediated immunosuppression [58]. Furthermore, MSCs can produce numerous immunosuppressive molecules, such as IL-6, PGE2, IDO, and IL-10, in response to inflammatory stimuli. PGE2 has been reported to mediate the MSC-mediated suppression of T cells, NK cells, and macrophages. Moreover, PGE2 has been found to act with IDO to alter the proliferation of T cells and NK cells [59]. In contrast, MSCs have come to be recognized as one type of adult stem cell actively participating in tissue repair by closely interacting with inflammatory cells and various other cell types [60]. Numerous reports have demonstrated that MSCs can release an array of growth and inhibitory factors, such as EGF, FGF, PDGF, and VEGF, and express several leukocyte chemokines, such as CXCL9, CCL2, CXCL10, and CXCL11. These factors provide an important microenvironment to activate adaptive immunity for lung repair [61]. Thus, the dual functions of MSCs may improve lung recovery after human CoV-induced ALI. Recently, MSCs was transplanted intravenously to enrolled patients with COVID-19 pneumonia. After treatment, the pulmonary function and symptoms of these patients were significantly improved. Meanwhile, the peripheral lymphocytes were increased, the C-reactive protein decreased, the level of TNF- was significantly decreased, and the overactivated cytokine-secreting immune cells disappeared. In addition, a group of regulatory DC cell population dramatically increased. Thus, the intravenous transplantation of MSCs was effective for treatment in patients with COVID-19 pneumonia [62, 63].

Stem cell therapies for treatment of influenza virus and coronavirus-induced lung injury. CoVs, coronavirus; MSCs, mesenchymal stem/stromal cells; LSCs, lung stem/progenitor cells; NK cells, natural killer cells; DC cells, dendritic cells

In addition, endogenous LSCs also play an important role in lung cell reconstitution after virus-induced ALI. In particular, TPR63+/KRT5+ airway BCs comprise approximately equal numbers of stem cells and committed precursors and give rise to differentiated luminal cells during steady state and epithelial repair after lung injury [44, 64]. Research has shown that KRT5+ cells repopulate damaged alveolar parenchyma following influenza virus infection [47]. However, there is still little evidence for the role of altered TPR63+/KRT5+ stem cells during lung injury repair caused by human CoVs.

In summary, exogenous MSCs may modulate human CoV-induced lung injury repair and regeneration through their immunoregulatory properties. These cells are capable of interacting with various types of immune cell, including neutrophils, macrophages, T cells, B cells, NK cells, and DCs. Furthermore, viral infections can activate endogenous LSCs to produce new lung cells and maintain lung function (Fig.1). Thus, we propose that MSCs and LSCs are two potential cell sources for treating human CoV-induced lung injury.

Read the original:
Stem cell therapy: a potential approach for treatment of influenza virus and coronavirus-induced acute lung injury - BMC Blogs Network

Posted in Stem Cell Research | Comments Off on Stem cell therapy: a potential approach for treatment of influenza virus and coronavirus-induced acute lung injury – BMC Blogs Network

Stem Cell Treatment Bala Cynwyd | Stem Cell Therapy …

Posted: May 25, 2020 at 8:44 am

Stem Cell Treatment in Philadelphia, PA

To treat joint and musculoskeletal system injuries, or if you suffer from chronic conditions affecting the joints or bones, there are options other than surgery to treat the issues. At World Wellness Health Institute, Dr. Daniel Lebowitz uses stem cell therapy on people living in and around Bala Cynwyd and Philadelphia, PA, who have chronic conditions and are in need of advanced treatment to heal injuries and relieve pain associated with injuries, arthritis, and other conditions.

Stem cell therapy is a cutting-edge treatment used in orthopedic injuries and other chronic conditions that affect the musculoskeletal system, such as arthritis and neck, back and joint pain. It is now a consideration for treatment instead of surgery. With stem cell therapy, many patients regain full function of the treated area without a lengthy recovery period, unlike with surgery. This type of stem cell therapy is FDA approved and takes stem cells from the patients own adipose tissue. This tissue (fat tissue) is rich in stem cells, primarily mesenchymalstem cells.

Stem cell injections are used primarily to relieve pain in the joints as they provide the following benefits:

With stem cell therapy, fat is harvested in a process called lipoaspiration. The targeted area is numbed with a local anesthetic, then a small needle just a tad larger than a hypodermic needle is injected into the skin to remove about 10 to 20ccs of adipose tissue (fat). Once the fat is removed, it is run through another process called sterile gravity method and combined with high-density PRP (platelet-rich plasma) and injected into the site where the pain occurs. Stem cells stimulate the healing process providing several functions. They can differentiate or even change into the type of cells needed, whether its a ligament, tendon, bone or cartilage, at the injection site to start healing. We prescribe oral anti-anxiety medicine and pain medicines that you can take prior to the procedure. If necessary, Dr. Lebowitz may use local anesthetic to numb the injection sites. You typically need only one treatment but, if necessary, a follow-up treatment may be performed several weeks later.

Much of the preparation for stem cell therapy involves determining if the patient is a good candidate for this type of treatment. It is necessary to stop taking any medications and/or supplements that may thin the blood at least one week prior to the treatment, as well as avoid smoking.

Stem cell therapy treatment is not a painful procedure, as oral pain relievers and local anesthetics are used to numb the treatment areas. It is okay to return to work immediately after the procedure, as well as participate in any activities. Most patients notice improvement after two weeks and continue to experience improvement over the next few months following the treatment.

Stem cell therapy is a relatively new procedure and varies in cost depending on the area to be treated, how many treatments may be required and if it is done at the same time another treatment is performed. We can discuss the cost with you during your consultation, in addition to going over our payment options. We do offerfinancingthrough CareCredit.

If you suffer from osteoarthritis or an injury to the knee, back, neck or other joint, stem cell therapy may be right for you. Stem cell therapy is an alternative to invasive surgery when the injured area is not fully collapsed.

During your consultation to determine if you are a candidate for stem cell therapy, it is necessary for Dr. Lebowitz to examine and evaluate the area of your pain, as well as determine the root cause of the pain. Additionally, he will go over your current health, medical history, and lifestyle in terms of diet, exercise, and more. If you have any questions, he answers them in detail so that you fully understand the treatment plan.

If you are experiencing pain in any joint or other part of the musculoskeletal system as a result of an injury or chronic health condition, stem cell therapy offered at World Wellness Health Institute may be the solution. Dr. Daniel Lebowitz offers several treatments for musculoskeletal therapy to residents in Bala Cynwyd, Philadelphia and the surrounding areas of Pennsylvania.Contact ustoday!*Individual results may vary

Read the original post:
Stem Cell Treatment Bala Cynwyd | Stem Cell Therapy ...

Posted in Pennsylvania Stem Cells | Comments Off on Stem Cell Treatment Bala Cynwyd | Stem Cell Therapy …

Gene therapy for follistatin mitigates systemic metabolic inflammation and post-traumatic arthritis in high-fat dietinduced obesity – Science Advances

Posted: May 13, 2020 at 8:52 pm

Abstract

Obesity-associated inflammation and loss of muscle function play critical roles in the development of osteoarthritis (OA); thus, therapies that target muscle tissue may provide novel approaches to restoring metabolic and biomechanical dysfunction associated with obesity. Follistatin (FST), a protein that binds myostatin and activin, may have the potential to enhance muscle formation while inhibiting inflammation. Here, we hypothesized that adeno-associated virus 9 (AAV9) delivery of FST enhances muscle formation and mitigates metabolic inflammation and knee OA caused by a high-fat diet in mice. AAV-mediated FST delivery exhibited decreased obesity-induced inflammatory adipokines and cytokines systemically and in the joint synovial fluid. Regardless of diet, mice receiving FST gene therapy were protected from post-traumatic OA and bone remodeling induced by joint injury. Together, these findings suggest that FST gene therapy may provide a multifactorial therapeutic approach for injury-induced OA and metabolic inflammation in obesity.

Osteoarthritis (OA) is a multifactorial family of diseases, characterized by cartilage degeneration, joint inflammation, and bone remodeling. Despite the broad impact of this condition, there are currently no disease-modifying drugs available for OA. Previous studies demonstrate that obesity and dietary fatty acids (FAs) play a critical role in the development of OA, and metabolic dysfunction secondary to obesity is likely to be a primary risk factor for OA (1), particularly following joint injury (2, 3). Furthermore, both obesity and OA are associated with a rapid loss of muscle integrity and strength (4), which may contribute directly and indirectly to the onset and progression of OA (5). However, the mechanisms linking obesity, muscle, and OA are not fully understood and appear to involve interactions among biomechanical, inflammatory, and metabolic factors (6). Therefore, strategies that focus on protecting muscle and mitigating metabolic inflammation may provide an attractive target for OA therapies in this context.

A few potential interventions, such as weight loss and exercise, have been proposed to reverse the metabolic dysfunction associated with obesity by improving the quantity or quality of skeletal muscle (7). Skeletal muscle mass is modulated by myostatin, a member of the transforming growth factor (TGF-) superfamily and a potent negative regulator of muscle growth (8), and myostatin is up-regulated in obesity and down-regulated by exercise (9). While exercise and weight loss are the first line of therapy for obesity and OA, several studies have shown difficulty in achieving long-term maintenance of weight loss or strength gain, particularly in frail or aging populations (10). Thus, targeted pharmacologic or genetic inhibition of muscle-regulatory molecules such as myostatin provides a promising approach to improving muscle metabolic health by increasing glucose tolerance and enhancing muscle mass in rodents and humans (8).

Follistatin (FST), a myostatin- and activin-binding protein, has been used as a therapy for several degenerative muscle diseases (11, 12), and loss of FST is associated with reduced muscle mass and prenatal death (13). In the context of OA, we hypothesize that FST delivery using a gene therapy approach has multifactorial therapeutic potential through its influence on muscle growth via inhibition of myostatin activity (14) as well as other members of the TGF- family. Moreover, FST has been reported to reduce the infiltration of inflammatory cells in the synovial membrane (15) and affect bone development (16), and pretreatment with FST has been shown to reduce the severity of carrageenan-induced arthritis (15). However, the potential for FST as an OA therapy has not been investigated, especially in exacerbating pathological conditions such as obesity. We hypothesized that overexpression of FST using a gene therapy approach will increase muscle mass and mitigate obesity-associated metabolic inflammation, as well as the progression of OA, in high-fat diet (HFD)induced obese mice. Mice fed an HFD were treated with a single dose of adeno-associated virus 9 (AAV9) to deliver FST or a green fluorescent protein (GFP) control, and the effects on systemic metabolic inflammation and post-traumatic OA were studied (fig. S1).

Dual-energy x-ray absorptiometry (DXA) imaging of mice at 26 weeks of age (Fig. 1A) showed significant effects of FST treatment on body composition. Control-diet, FST-treated mice (i.e., Control-FST mice) exhibited significantly lower body fat percentages, but were significantly heavier than mice treated with a GFP control vector (Control-GFP mice) (Fig. 1B), indicating that increased muscle mass rather than fat was developed with FST. With an HFD, control mice (HFD-GFP mice) showed significant increases in weight and body fat percentage that were ameliorated by FST overexpression (HFD-FST mice).

(A) DXA images of mice at 26 weeks of age. (B) DXA measurements of body fat percentage and bone mineral density (BMD; 26 weeks) and body weight measurements over time. (C) Serum levels for adipokines (insulin, leptin, resistin, and C-peptide) at 28 weeks. (D) Metabolite levels for glucose, triglycerides, cholesterol, and FFAs at 28 weeks. (E) Serum levels for cytokines (IL-1, IL-1, MCP-1, and VEGF) at 28 weeks. (F) Fluorescence microscopy images of visceral adipose tissue with CD11b:Alexa Fluor 488 (green), CD11c:phycoerythrin (PE) (red), and 4,6-diamidino-2-phenylindole (DAPI; blue). Scale bars, 100 m. Data are presented as mean SEM; n = 8 to 10; two-way analysis of variance (ANOVA), P < 0.05. Groups not sharing the same letter are significantly different with Tukey post hoc analysis. For IL-1 and VEGF, P < 0.05 for diet effect and AAV effect. For MCP-1, P < 0.05 for diet effect.

In the HFD group, overexpression of FST significantly decreased serum levels of several adipokines including insulin, leptin, resistin, and C-peptide as compared to GFP-treated mice (Fig. 1C). HFD-FST mice also had significantly lower serum levels of glucose, triglycerides, cholesterol, and free FAs (FFAs) (Fig. 1D), as well as the inflammatory cytokine interleukin-1 (IL-1) (Fig. 1E) when compared to HFD-GFP mice. For both dietary groups, AAV-FST delivery significantly increased circulating levels of vascular endothelial growth factor (VEGF) while significantly decreasing IL-1 levels. Furthermore, obesity-induced inflammation in adipose tissue was verified by the presence of CD11b+CD11c+ M1 pro-inflammatory macrophages or dendritic cells (Fig. 1F).

To determine whether FST gene therapy can mitigate injury-induced OA, mice underwent surgery for destabilization of the medial meniscus (DMM) and were sacrificed 12 weeks after surgery. Cartilage degeneration was significantly reduced in DMM joints of the mice receiving FST gene therapy in both dietary groups (Fig. 2, A and C) when compared to GFP controls. FST overexpression also significantly decreased joint synovitis (Fig. 2, B and D) when compared to GFP controls. To evaluate the local influence of pro-inflammatory cytokines to joint degeneration and inflammation, synovial fluid (SF) was harvested from surgical and ipsilateral nonsurgical limbs and analyzed using a multiplexed array. The DMM joints from mice with FST overexpression exhibited a trend toward lower levels of pro-inflammatory cytokines, including IL-1, IL-1, and IL-6, and a higher level of interferon- (IFN-)induced protein (IP-10) in the SF of DMM joints as compared to contralateral controls (Fig. 2E).

(A) Histologic analysis of OA severity via Safranin O (glycosaminoglycans) and fast green (bone and tendon) staining of DMM-operated joints. (B) Histology [hematoxylin and eosin (H&E) staining] of the medial femoral condyle of DMM-operated joints. Thickened synovium (S) from HFD mice with a high density of infiltrated cells was observed (arrows). (C) Modified Mankin scores compared within the diet. (D) Synovitis scores compared within the diet. (E) Levels of proinflammatory cytokines in the SF compared within the diet. (F) Hot plate latency time and sensitivity to cold plate exposure, as measured using the number of jumps in 30 s, both for non-operated algometry measurements of pain sensitivity compared within the diet. Data are presented as mean SEM; n = 5 to 10 mice per group; two-way ANOVA, P < 0.05. Groups not sharing the same letter are significantly different with Tukey post hoc analysis.

To investigate the effect of FST on pain sensitivity in OA, animals were subjected to a variety of pain measurements including hot plate, cold plate, and algometry. Obesity increased heat withdrawal latency, which was rescued by FST overexpression (Fig. 2F). Cold sensitivity trended lower with obesity, and because no significant differences in heat withdrawal latency were found with surgery (fig. S2), no cold sensitivity was measured after surgery. We found that FST treatment protected HFD animals from mechanical algesia at the knee receiving DMM surgery, while Control-diet DMM groups demonstrated increased pain sensitivity following joint injury.

A bilinear regression model was used to elucidate the relationship among OA severity, biomechanical factors, and metabolic factors (table S1). Factors significantly correlated with OA were then selected for multivariate regression (Table 1). Both multivariate regression models revealed serum tumor necrosis factor- (TNF-) levels as a major predictor of OA severity.

, standardized coefficient. ***P < 0.001.

We analyzed the effects of FST treatment on muscle structure and mass, and performance measures were conducted on mice in both dietary groups. Both Control-FST and HFD-FST limbs exhibited visibly larger muscles compared to both AAV-GFP groups (Fig. 3A). In addition, the muscle masses of tibialis anterior (TA), gastrocnemius, and quadriceps increased significantly with FST treatment (Fig. 3B). Western blot analysis confirmed an increase in FST expression in the muscle at the protein level in FST-treated groups compared to GFP-treated animals in Control and HFD groups (Fig. 3C). Immunofluorescence labeling showed increased expression of FST in muscle (Fig. 3D) and adipose tissue (Fig. 3E) of the AAV-FST mice, with little or no expression of FST in control groups.

(A) Photographic images and (B) measured mass of tibialis anterior (TA), gastrocnemius (GAS), and quadriceps (QUAD) muscles; n = 8, diet and AAV effects both P < 0.05. (C) Western blot showing positive bands of FST protein only in FST-treated muscles, with -actin as a loading control. Immunolabeling of (D) GAS muscle and (E) adipose tissue showing increased expression of FST, particularly in skeletal muscle. (F) H&E-stained sections of GAS muscles were measured for (G) mean myofiber diameter; n = 100 from four mice per group, diet, and AAV effects; both P < 0.05. (H) Oil Red O staining was analyzed for (I) optical density values of FAs; n = 6. (J) Second-harmonic generation imaging of collagen in TA sections was quantified for intensity; n = 6. (K) Western blotting showing the level of phosphorylation markers of protein synthesis in GAS muscle. (L) Functional analysis of grip strength and treadmill time to exhaustion; n = 10. Data are presented as mean SEM; two-way ANOVA, P < 0.05. Groups not sharing the same letter are significantly different with Tukey post hoc analysis. Photo credit: Ruhang Tang, Washington University.

To determine whether the increases in muscle mass reflected muscle hypertrophy, gastrocnemius muscle fiber diameter was measured in H&E-stained sections (Fig. 3F) at 28 weeks of age. Mice with FST overexpression exhibited increased fiber diameter (i.e., increased muscle hypertrophy) relative to the GFP-expressing mice in both diet treatments (Fig. 3G). Oil Red O staining was used to determine the accumulation of neutral lipids in muscle (Fig. 3H). We found that HFD-FST mice were protected from lipid accumulation in muscles compared to HFD-GFP mice (Fig. 3I). Second-harmonic generation imaging confirmed the presence of increased collagen content in the muscles of HFD mice, which was prevented by FST gene therapy (Fig. 3J). We also examined the expression and phosphorylation levels of the key proteins responsible for insulin signaling in muscles. We observed increased phosphorylation of AktS473, S6KT389, and S6RP-S235/2369 and higher expression of peroxisome proliferatoractivated receptor coactivator 1- (Pgc1-) in muscles from FST mice compared to GFP mice, regardless of diet (Fig. 3K). In addition to the improvements in muscle structure with HFD, FST-overexpressing mice also showed improved function, including higher grip strength and increased treadmill running endurance (Fig. 3L), compared to GFP mice.

Because FST has the potential to influence cardiac muscle and skeletal muscle, we performed a detailed evaluation on the effect of FST overexpression on cardiac function. Echocardiography and short-axis images were collected to visualize the left ventricle (LV) movement during diastole and systole (fig. S3A). While the Control-FST mice had comparable LV mass (LVM) and left ventricular posterior wall dimensions (LVPWD) with Control-GFP mice (fig. S3, B and C), the HFD-FST mice have significantly decreased LVM and trend toward decreased LVPWD compared to HFD-GFP. Regardless of the diet treatments, FST overexpression enhanced the rate of heart weight/body weight (fig. S3D). Although Control-FST mice had slightly increased dimensions of the interventricular septum at diastole (IVSd) compared to Control-GFP (fig. S3E), there was significantly lower IVSd in HFD-FST compared to HFD-GFP. In addition, we found no difference in fractional shortening among all groups (fig. S3F). Last, transmitral blood flow was investigated using pulse Doppler. While there was no difference in iso-volumetric relaxation time (IVRT) in Control groups, HFD-FST mice had a moderate decrease in IVRT compared to HFD-GFP (fig. S3G). Overall, FST treatment mitigated the changes in diastolic dysfunction and improved the cardiac relaxation caused by HFD.

DXA demonstrated that FST gene therapy improved bone mineral density (BMD) in HFD compared to other groups (Fig. 1B). To determine the effects of injury, diet intervention, and overexpression of FST on bone morphology, knee joints were evaluated by microcomputed tomography (microCT) (Fig. 4A). The presence of heterotopic ossification was observed throughout the GFP knee joints, whereas FST groups demonstrated a reduction or an absence of heterotopic ossification. FST overexpression significantly increased the ratio of bone volume to total volume (BV/TV), BMD, and trabecular number (Tb.N) of the tibial plateau in animals, regardless of diet treatment (Fig. 4B). Joint injury generally decreased bone parameters in the tibial plateau, particularly in Control-diet mice. In the femoral condyle, BV/TV and Tb.N were significantly increased in mice with FST overexpression in both diet types, while BMD was significantly higher in HFD-FST compared to HFD-GFP mice (Fig. 4B). Furthermore, AAV-FST delivery significantly increased trabecular thickness (Tb.Th) and decreased trabecular space (Tb.Sp) in the femoral condyle of HFD-FST compared to HFD-GFP animals (fig. S4).

(A) Three-dimensional (3D) reconstruction of microCT images of non-operated and DMM-operated knees. (B) Tibial plateau (TP) and femoral condyle (FC) regional analyses of trabecular bone fraction bone volume (BV/TV), BMD, and trabecular number (Tb.N). Data are presented as mean SEM; n = 8 to 19 mice per group; two-way ANOVA. (C) 3D microCT reconstruction of metaphysis region of DMM-operated joints. (D) Analysis of metaphysis BV/TV, Tb.N, and BMD. (E) 3D microCT reconstruction of cortical region of DMM-operated joints. (F) Analysis of cortical cross-sectional thickness (Ct.Cs.Th), polar moment of inertia (MMI), and tissue mineral density (TMD). (D and F) Data are presented as mean SEM; n = 8 to 19 mice per group; Mann-Whitney U test, *P < 0.05.

Further microCT analysis was conducted on the trabecular (Fig. 4C) and cortical (Fig. 4E) areas of the metaphyses. FST gene therapy significantly increased BV/TV, Tb.N, and BMD in the metaphyses regardless of the diet (Fig. 4D). Furthermore, FST delivery significantly increased the cortical cross-sectional thickness (Ct.Cs.Th) and polar moment of inertia (MMI) of mice on both diet types, as well as tissue mineral density (TMD) of cortical bones of mice fed control diet (Fig. 4F).

To elucidate the possible mechanisms by which FST mitigates inflammation, we examined the browning/beiging process in subcutaneous adipose tissue (SAT) with immunohistochemistry (Fig. 5A). Here, we found that key proteins expressed mainly in brown adipose tissue (BAT) (PGC-1, PRDM16, thermogenesis marker UCP-1, and beige adipocyte marker CD137) were up-regulated in SAT of the mice with FST overexpression (Fig. 5B). Increasing evidence suggests that an impaired mitochondrial oxidative phosphorylation (OXPHOS) system in white adipocytes is a hallmark of obesity-associated inflammation (17). Therefore, we further examined the mitochondrial respiratory system in SAT. HFD reduced the amount of OXPHOS complex subunits (Fig. 5C). We found that proteins involved in OXPHOS, including subunits of complexes I, II, and III of mitochondria OXPHOS complex, were significantly up-regulated in AAV-FSToverexpressing animals compared to AAV-GFP mice (Fig. 5D).

(A) Immunohistochemistry of UCP-1 expression in SAT. Scale bar, 50 m. (B) Western blotting of SAT for key proteins expressed in BAT, with -actin as a loading control. (C) Western blot analysis of mitochondria lysates from SAT for OXPHOS proteins using antibodies against subunits of complexes I, II, III, and IV and adenosine triphosphate (ATP) synthase. (D) Change of densitometry quantification normalized to the average FST level of each OXPHOS subunit. Data are presented as mean SEM; n = 3. *P < 0.05, t test comparison within each pair.

Our findings demonstrate that a single injection of AAV-mediated FST gene therapy ameliorated systemic metabolic dysfunction and mitigated OA-associated cartilage degeneration, synovial inflammation, and bone remodeling occurring with joint injury and an HFD. Of note, the beneficial effects were observed across multiple tissues of the joint organ system, underscoring the value of this potential treatment strategy. The mechanisms by which obesity and an HFD increase OA severity are complex and multifactorial, involving increased systemic metabolic inflammation, joint instability and loss of muscle strength, and synergistic interactions between local and systemic cytokines (4, 6). In this regard, the therapeutic consequences of FST gene therapy also appear to be multifactorial, involving both direct and indirect effects such as increased muscle mass and metabolic activity to counter caloric intake and metabolic dysfunction resulting from an HFD while also promoting adipose tissue browning. Furthermore, FST may also serve as a direct inhibitor of growth factors in the TGF- family that may be involved in joint degeneration (18).

FST gene therapy showed a myriad of notable beneficial effects on joint degeneration following joint injury while mitigating HFD-induced obesity. These data also indirectly implicate the critical role of muscle integrity in the onset and progression of post-traumatic OA in this model. It is important to note that FST gene therapy mitigated many of the key negative phenotypic changes previously associated with obesity and OA, including cartilage structural changes as well as bone remodeling, synovitis, muscle fibrosis, and increased pain, as compared to GFP controls. To minimize the number of animals used, we did not perform additional controls with no AAV delivery; however, our GFP controls showed similar OA changes as observed in our previous studies, which did not involve any gene delivery (2). Mechanistically, FST restored to control levels a number of OA-associated cytokines and adipokines in the serum and the SF. While the direct effects of FST on chondrocytes remains to be determined, FST has been shown to serve as a regulator of the endochondral ossification process during development (19), which may also play a role in OA (20). Furthermore, previous studies have shown that a 2-week FST treatment of mouse joints is beneficial in reducing infiltration of inflammatory cells into the synovial membrane (15). Our findings suggest that FST delivery in skeletally mature mice, preceding obesity-induced OA changes, substantially reduces the probability of tissue damage.

It is well recognized that FST can inhibit the activity of myostatin and activin, both of which are up-regulated in obesity-related modalities and are involved in muscle atrophy, tissue fibrosis, and inflammation (21). Consistent with previous studies, our results show that FST antagonizes the negative regulation of myostatin in muscle growth, reducing adipose tissue content in animals. Our observation that FST overexpression decreased inflammation at both serum systemic and local joint inflammation may provide mechanistic insights into our findings of mitigated OA severity in HFD-fed mice. Our statistical analysis implicated serum TNF- levels as a major factor in OA severity, consistent with previous studies linking obesity and OA in mice (22). Although the precise molecular mechanisms of FST in modulating inflammation remain unclear, some studies postulate that FST may act like acute-phase protein in lipopolysaccharide-induced inflammation (23).

In addition to these effects of skeletal muscle, we found that FST gene therapy normalized many of the deleterious changes of an HFD on cardiac function without causing hypertrophy. These findings are consistent with previous studies showing that, during the process of aging, mice with myostatin knockout had an enhanced cardiac stress response (24). Furthermore, FST has been shown to regulate activin-induced cardiomyocyte apoptosis (1). In the context of this study, it is also important to note that OA has been shown to be a serious risk factor for progression of cardiovascular disease (25), and severity of OA disability is associated with significant increases in all-cause mortality and cardiovascular events (26).

FST gene therapy also rescued diet- and injury-induced bone remodeling in the femoral condyle, as well as the tibial plateau, metaphysis, and cortical bone of the tibia, suggesting a protective effect of FST on bone homeostasis of mice receiving an HFD. FST is a known inhibitor of bone morphogenetic proteins (BMPs), and thus, the interaction between the two proteins plays an essential role during bone development and remodeling. For example, mice grown with FST overexpression via global knock-in exhibited an impaired bone structure (27). However, in adult diabetic mice, FST was shown to accelerate bone regeneration by inhibiting myostatin-induced osteoclastogenesis (28). Furthermore, it has been reported that FST down-regulates BMP2-driven osteoclast activation (29). Therefore, the protective role of FST on obesity-associated bone remodeling, at least in part, may result from the neutralizing capacity of FST on myostatin in obesity. In addition, improvement in bone quality in FST mice may be explained by their enhanced muscle mass and strength, as muscle mass can dominate the process of skeletal adaptation, and conversely, muscle loss correlates with reduced bone quality (30).

Our results show that FST delivery mitigated pain sensitivity in OA joints, a critical aspect of clinical OA. Obesity and OA are associated with both chronic pain and pain sensitization (31), but it is important to note that structure and pain can be uncoupled in OA (32), necessitating the measurement of both behavioral and structural outcomes. Of note, FST treatment protected only HFD animals from mechanical algesia at the knee post-DMM surgery and also rescued animals from pain sensitization induced by HFD in both the DMM and nonsurgical limb. The mitigation in pain sensitivity observed here with FST treatment may also be partially attributed to the antagonistic effect of FST on activin signaling. In addition to its role in promoting tissue fibrosis, activin A has been shown to regulate nociception in a manner dependent on the route of injection (33, 34). It has been shown that activin can sensitize the transient receptor potential vanilloid 1 (TRPV1) channel, leading to acute thermal hyperalgesia (33). However, it is also possible that activin may induce pain indirectly, for example, by triggering neuroinflammation (35), which could lead to sensitization of nociceptors.

The earliest detectable abnormalities in subjects at risk for developing obesity and type 2 diabetes are muscle loss and accumulation of excess lipids in skeletal muscles (4, 36), accompanied by impairments in nuclear-encoded mitochondrial gene expression and OXPHOS capacity of muscle and adipose tissues (17). PGC-1 activates mitochondrial biogenesis and increases OXPHOS by increasing the expression of the transcription factors necessary for mitochondrial DNA replication (37). We demonstrated that FST delivery can rescue low levels of OXPHOS in HFD mice by increasing expression PGC-1 (Fig. 3H). It has been reported that high-fat feeding results in decreased PGC-1 and mitochondrial gene expression in skeletal muscles, while exercise increases the expression of PGC-1 in both human and rodent muscles (38, 39). Although the precise molecular mechanism by which FST promotes PGC-1 expression has not been established, the infusion of lipids decreases expression of PGC-1 and nuclear-encoded mitochondrial genes in muscles (40). Thus, decreased lipid accumulation in muscle by FST overexpression may provide a plausible explanation for the restored PGC-1 in the FST mice. These findings were further confirmed by the metabolic profile, showing reduced serum levels of triglycerides, glucose, FFAs, and cholesterol (Fig. 1D), and are consistent with previous studies, demonstrating that muscles with high numbers of mitochondria and oxidative capacity (i.e., type 1 muscles with high levels of PGC-1 expression) are protected from damage due to an HFD (4).

In addition, we found increased phosphorylation of protein kinase B (Akt) on Ser473 in the skeletal muscle of FST-treated mice as compared to untreated HFD counterparts (Fig. 3K), consistent with restoration of a normal insulin response. A number of studies have demonstrated that the serine-threonine protein kinase Akt1 is a critical regulator of cellular hypertrophy, organ size, and insulin signaling (41). Muscle hypertrophy is stimulated both in vitro and in vivo by the expression of constitutively active Akt1 (42, 43). Furthermore, it has been demonstrated that constitutively active Akt1 also promotes the production of VEGF (44).

BAT is thought to be involved in thermogenesis rather than energy storage. BAT is characterized by a number of small multilocular adipocytes containing a large number of mitochondria. The process in which white adipose tissue (WAT) becomes BAT, called beiging or browning, is postulated to be protective in obesity-related inflammation, as an increase in BAT content positively correlates with increased triglyceride clearance, normalized glucose level, and reduced inflammation. Our study shows that AAV-mediated FST delivery serves as a very promising approach to induce beiging of WAT in obesity. A recent study demonstrated that transgenic mice overexpressing FST exhibited an increasing amount of BAT and beiging in subcutaneous WAT with increased expression of key BAT-related markers including UCP-1 and PRDM16 (45). In agreement with previous reports, our data show that Ucp1, Prdm16, Pgc1a, and Cd167 are significantly up-regulated in SAT of mice overexpressing FST in both dietary interventions. FST has been recently demonstrated to play a crucial role in modulating obesity-induced WAT expansion by inhibiting TGF-/myostatin signaling and thus promoting overexpression of these key thermogenesis-related genes. Together, these findings suggest that the observed reduction in systemic inflammation in our model may be partially explained by FST-mediated increased process of browning/beiging.

In conclusion, we show that a single injection of AAV-mediated FST, administered after several weeks of HFD feeding, mitigated the severity of OA following joint injury, and improved muscle performance as well as induced beiging of WAT, which together appeared to decrease obesity-associated metabolic inflammation. These findings provide a controlled model for further examining the differential contributions of biomechanical and metabolic factors to the progression of OA with obesity or HFD. As AAV gene therapy shows an excellent safety profile and is currently in clinical trials for a number of conditions, such an approach may allow the development of therapeutic strategies not only for OA but also, more broadly, for obesity and associated metabolic conditions, including diseases of muscle wasting.

All experimental procedures were approved by and conducted in accordance with the Institutional Animal Care and Use Committee guidelines of Washington University in Saint Louis. The overall timeline of the study is shown in fig. S1A. Beginning at 5 weeks of age, C57BL/6J mice (The Jackson Laboratory) were fed either Control or 60% HFD (Research Diets, D12492). At 9 week of age, mice received AAV9-mediated FST or GFP gene delivery via tail vein injection. A total of 64 mice with 16 mice per dietary group per AAV group were used. DMM was used to induce knee OA in the left hind limbs of the mice at the age of 16 weeks. The non-operated right knees were used as contralateral controls. Several behavioral activities were measured during the course of the study. Mice were sacrificed at 28 weeks of age to evaluate OA severity, joint inflammation, and joint bone remodeling.

Mice were weighed biweekly. The body fat content and BMD of the mice were measured using a DXA (Lunar PIXImus) at 14 and 26 weeks of age, respectively.

Complementary DNA synthesis for mouse FST was performed by reverse transcriptase in a reverse transcription quantitative polymerase chain reaction (RT-qPCR) ( Invitrogen) mixed with mRNAs isolated from the ovary tissues of C57BL/6J mouse. The PCR product was cloned into the AAV9-vector plasmid (pTR-UF-12.1) under the transcriptional control of the chicken -actin (CAG) promoter including cytomegalovirus (CMV) enhancers and a large synthetic intron (fig. S1B). Recombinant viral vector stocks were produced at Hope Center Viral Vectors Core (Washington University, St. Louis) according to the plasmid cotransfection method and suspension culture. Viral particles were purified and concentrated. The purity of AAV-FST and AAV-GFP was evaluated by SDSpolyacrylamide gel electrophoresis (PAGE) and stained by Coomassie blue. The results showed that the AAV protein components in 5 1011 vector genomes (vg) are only stained in three major protein bands: VR1, 82 kDa; VR2, 72 kDa; and VR3, 62 kDa. Vector titers were determined by the DNA dot-blot and PCR methods and were in the range of 5 1012 to 1.5 1013 vector copies/ml. AAV was delivered at a final dose of 5 1011 vg per mouse by intravenous tail injection under red light illumination at 9 weeks of age. This dose was determined on the basis of our previous studies showing that AAV9-FST gene delivery by this route resulted in a doubling of muscle mass at a dose of 2.5 1011 vg in 4-week-old mice or at 5 1011 vg in 8-week-old mice (46).

At 16 weeks of age, mice underwent surgery for the DMM to induce knee OA in the left hindlimb as previously described (2). Briefly, anesthetized mice were placed on a custom-designed device, which positioned their hindlimbs in 90 flexion. The medial side of the joint capsule was opened, and the medial meniscotibial ligament was transected. The joint capsule and subcutaneous layer of the skin were closed with resorbable sutures.

Mice were sacrificed at 28 weeks of age, and changes in joint structure and morphology were assessed using histology. Both hindlimbs were harvested and fixed in 10% neutral-buffered formalin (NBF). Limbs were then decalcified in Cal-Ex solution (Fisher Scientific, Pittsburgh, PA, USA), dehydrated, and embedded in paraffin. The joint was sectioned in the coronal plane at a thickness of 8 m. Joint sections were stained with hematoxylin, fast green, and Safranin O to determine OA severity. Three blinded graders then assessed sections for degenerative changes of the joint using a modified Mankin scoring system (2). Briefly, this scoring system measures several aspects of OA progression (cartilage structure, cell distribution, integrity of tidemark, and subchondral bone) in four joint compartments (medial tibial plateau, medial femoral condyle, lateral tibial plateau, and lateral femoral condyle), which are summed to provide a semiquantitative measure of the severity of joint damage. To assess the extent of synovitis, sections were stained with H&E to analyze infiltrated cells and synovial structure. Three independent blinded graders scored joint sections for synovitis by evaluating synovial cell hyperplasia, thickness of synovial membrane, and inflammation in subsynovial regions in four joint compartments, which were summed to provide a semiquantitative measure of the severity of joint synovitis (2). Scores for the whole joint were averaged among graders.

Serum and SF from the DMM and contralateral control limbs were collected, as described previously (2). For cytokine and adipokine levels in the serum and SF fluid, a multiplexed bead assay (Discovery Luminex 31-Plex, Eve Technologies, Calgary, AB, Canada) was used to determine the concentration of Eotaxin, granulocyte colony-stimulating factor (G-CSF), granulocyte-macrophage CSF (GM-CSF), IFN-, IL-1, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-9, IL-10, IL-12 (p40), IL-12 (p70), IL-13, IL-15, IL-17A, IP-10, keratinocyte chemoattractant (KC), leukemia inhibitory factor (LIF), liposaccharide-induced (LIX), monocyte chemoattractant protein-1 (MCP-1), M-CSF, monokine induced by gamma interferon (MIG), macrophage inflammatory protein1 (MIP-1), MIP-1, MIP-2, RANTES, TNF-, and VEGF. A different kit (Mouse Metabolic Array) was used to measure levels for amylin, C-peptide, insulinotropic polypeptide (GIP), glucagon-like peptide-1 (GLP-1), ghrelin, glucagon, insulin, leptin, protein phosphatase (PP), peptide yy (PYY), and resistin. Missing values were imputed using the lowest detectable value for each analyte.

Muscles were cryopreserved by incubation with 2-methylbutane in a steel beaker using liquid nitrogen for 30 s, cryoembedded, and cryosectioned at 8 m thickness. Tissue sections were stained following standard H&E protocol. Photomicrographs of skeletal muscle fiber were imaged under brightfield (VS120, Olympus). Muscle slides fixed in 3.7% formaldehyde were stained with 0.3% Oil Red O (in 36% triethyl phosphate) for 30 min. Images were taken in brightfield (VS120, Olympus). The relative concentration of lipid was determined by extracting the Oil Red O with isopropanol in equally sized muscle sections and quantifying the OD500 (optical density at 500 nm) in a 96-well plate.

To determine spatial expression of FST in different tissues, cryosections of gastrocnemius muscles and adipose tissue were immunolabeled for FST. Tissue sections were fixed in 1.5% paraformaldehyde solution, and primary anti-FST antibody (R&D Systems, AF-669, 1:50) was incubated overnight at 4C after blocking with 2.5% horse serum (Vector Laboratories), followed by labeling with a secondary antibody (Alexa Fluor 488, Invitrogen, A11055) and with 4,6-diamidino-2-phenylindole (DAPI) for cell nuclei. Sections were imaged using fluorescence microscopy.

Second-harmonic generation images of TA were obtained from unstained slices using backscatter signal from an LSM 880 confocal microscope (Zeiss) with Ti:sapphire laser tuned to 820 nm (Coherent). The resulting image intensity was analyzed using ImageJ software.

To measure bone structural and morphological changes, intact hindlimbs were scanned by microCT (SkyScan 1176, Bruker) with an 18-m isotropic voxel resolution (455 A, 700-ms integration time, and four-frame averaging). A 0.5-mm aluminum filter was used to reduce the effects of beam hardening. Images were reconstructed using NRecon software (with 10% beam hardening and 20 ring artifact corrections). Subchondral/trabecular and cortical bone regions were segmented using CTAn automatic thresholding software. Tibial epiphysis was selected using the subchondral plate and growth plate as references. Tibial metaphysis was defined as the 1-mm region directly below the growth plate. The cortical bone analysis was performed in the mid-shaft (4 mm below the growth plate with a height of 1 mm). Hydroxyapatite calibration phantoms were used to calibrate bone density values (mg/cm3).

Fresh visceral adipose tissues were collected, frozen in optimal cutting temperature compound (OCT), and cryosectioned at 5-m thickness. Tissue slides were then acetone-fixed followed by incubation with Fc receptor blocking in 2.5% goat serum (Vector Laboratories) and incubation with primary antibodies cocktail containing anti-CD11b:Alexa Fluor 488 and CD11c:phycoerythrin (PE) (BioLegend). Nuclei were stained with DAPI. Samples were imaged using fluorescence microscopy (VS120, Olympus).

Adipose tissues were fixed in 10% NBF, paraffin-embedded, and cut into 5-m sections. Sections were deparaffinized, rehydrated, and stained with H&E. Immunohistochemistry was performed by incubating sections (n = 5 per each group) with the primary antibody (antimUCP-1, U6382, Sigma), followed by a secondary antibody conjugated with horseradish peroxidase (HRP). Chromogenic substrate 3,3-diaminobenzidine (DAB) was used to develop color. Counterstaining was performed with Harris hematoxylin. Sections were examined under brightfield (VS120, Olympus).

Proteins of the muscle or fat tissue were extracted using lysis buffer containing 1% Triton X-100, 20 mM tris-HCl (pH 7.5), 150 mM NaCl, 1 mm EDTA, 5 mM NaF, 2.5 mM sodium pyrophosphate, 1 mM -glycerophosphate, 1 mM Na3VO4, leupeptin (1 g ml1), 0.1 mM phenylmethylsulfonyl fluoride, and a cocktail of protease inhibitors (Sigma, St. Louis, MO, USA, catalog no. P0044). Protein concentrations were measured with Quick Start Bradford Dye Reagent (Bio-Rad). Twenty micrograms of each sample was separated in SDS-PAGE gels with prestained molecular weight markers (Bio-Rad). Proteins were wet-transferred to polyvinylidene fluoride membranes. After incubating for 1.5 hours with a buffer containing 5% nonfat milk (Bio-Rad #170-6404) at room temperature in 10 mM tris-HCl (pH 7.5), 100 mM NaCl, and 0.1% Tween 20 (TBST), membranes were further incubated overnight at 4C with antiUCP-1 rabbit polyclonal antibody (1:500, Sigma, U6382), anti-PRDM16 rabbit antibody (Abcam, ab106410), anti-CD137 rabbit polyclonal antibody (1:1000, Abcam, ab203391), total OXPHOS rodent western blot (WB) antibodies (Abcam, ab110413), anti-actin (Cell Signaling Technology, 13E5) rabbit monoclonal antibody (Cell Signaling Technology, 4970), followed by HRP-conjugated secondary antibody incubation for 30 min. A chemiluminescent detection substrate (Clarity, Western ECL) was applied, and the membranes were developed (iBrightCL1000).

The effects of HFD and FST gene therapy on thermal hyperalgesia were examined at 15 weeks of age. Mice were acclimatized to all equipment 1 day before the onset of testing, as well as a minimum of 30 min before conducting each test. Thermal pain tests were measured in a room set to 25C. Peripheral thermal sensitivity was determined using a hot/cold analgesia meter (Harvard Apparatus, Holliston, MA, USA). For hot plate testing, the analgesia meter was set to 55C. To prevent tissue damage, a maximum cutoff time of 20 s was established a priori, at which time an animal would be removed from the plate in the absence of pain response, defined as paw withdrawal or licking. Animals were tested in the same order three times, allowing each animal to have a minimum of 30 min between tests. The analgesia meter was cleaned with 70% ethanol between trials. The average of the three tests was reported per animal. To evaluate tolerance to cold, the analgesia meter was set to 0C. After 1-hour rest, animals were tested for sensitivity to cold over a single 30-s exposure. The number of jumps counted per animal was averaged within each group and compared between groups.

Pressure-pain tests were conducted at the knee using a Small Animal Algometer (SMALGO, Bioseb, Pinellas Park, FL, USA). Surgical and nonsurgical animals were evaluated over serial trials on the lateral aspect of the experimental and contralateral knee joints. The average of three trials per limb was calculated for each limb. Within each group, the pain threshold of the DMM limb versus non-operated limb was compared using a t test run on absolute values of mechanical pain sensitivity for each limb, P 0.05.

To assess the effect of HFD and AAV-FST treatments on neuromuscular function, treadmill running to exhaustion (EXER3, Columbus Instruments) was performed at 15 m/min, with 5 inclination angle on the mice 4 months after gene delivery. Treadmill times were averaged within groups and compared between groups.

Forelimb grip strength was measured using Chatillon DFE Digital Force Gauge (Johnson Scale Co.) for front limb strength of the animals. Each mouse was tested five times, with a resting period of 90 s between each test. Grip strength measurements were averaged within groups and compared between groups.

Cardiac function of the mice was examined at 6 months of age (n = 3) using echocardiography (Vevo 2100 High-Resolution In Vivo Imaging System, VisualSonics). Short-axis images were taken to view the LV movement during diastole and systole. Transmitral blood flow was observed with pulse Doppler. All data and images were performed by a blinded examiner and analyzed with an Advanced Cardiovascular Package Software (VisualSonics).

Detailed statistical analyses are described in methods of each measurement and its corresponding figure captions. Analyses were performed using GraphPad Prism, with significance reported at the 95% confidence level.

This is an open-access article distributed under the terms of the Creative Commons Attribution-NonCommercial license, which permits use, distribution, and reproduction in any medium, so long as the resultant use is not for commercial advantage and provided the original work is properly cited.

Acknowledgments: Funding: This study was supported, in part, by NIH grants AR50245, AR48852, AG15768, AR48182, AG46927, AR073752, OD10707, AR060719, AR074992, and AR75899; the Arthritis Foundation; and the Nancy Taylor Foundation for Chronic Diseases. Author contributions: R.T. and F.G. developed the concept of the study; R.T., N.S.H., C.-L.W., K.H.C., and Y.-R.C. collected and analyzed data; S.J.O. analyzed data; and all authors contributed to the writing of 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.

View original post here:
Gene therapy for follistatin mitigates systemic metabolic inflammation and post-traumatic arthritis in high-fat dietinduced obesity - Science Advances

Posted in Gene therapy | Comments Off on Gene therapy for follistatin mitigates systemic metabolic inflammation and post-traumatic arthritis in high-fat dietinduced obesity – Science Advances