Category Archives: Adult Stem Cells

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

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

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

What It’s Like to Grow a ‘Mini-Brain’ From Your Own Cells – Discover Magazine

This story appeared in the June 2020 issue as "Out of His Mind."Subscribeto Discover magazine for more stories like this.

It isnt every day that writer Philip Ball is asked to contribute to scientific experiments typically, hes reporting on them instead. But in 2015, scientists from a London-based project called Created Out of Mind asked Ball and a few other creatives if theyd participate in their work and then reflect on the experience. He agreed.

Ball first gave a sample, made up of cells from his skin, which was reprogrammed into stem cells that were subsequently grown into a brain organoid or, as the research team called it, a miniature brain in a dish. These futuristic-sounding specimens arent literal brains, and they dont look it, either. The round, cream-colored clusters of neurons simply resemble little blobs. Theyre not conscious. But they can be studied to better understand brain development such as where and when certain proteins misfold, which can signal whether a person will develop dementia.

This stained microscope image shows cells growing in a cross-section of a brain organoid, with neurons visible in red. (Credit: Chris Lovejoy and Selina Wray)

The researchers then asked Ball to reflect on his experience through writing. What started as a few blogs for Created Out of Mind eventually bloomed into a book, How to Grow a Human. Here, Ball recounts his time spent with his mini brain, and how the process gave him new insight into the future of lab-grown life.

Id pop into the lab from time to time and see how things were going. At one point, I stopped by and one of the researchers told me that the organoids werent looking great for some reason, they werent growing as they thought they might. We werent sure quite how they were going to turn out, and in the end many of them died before they could fully form.

Philip Ball (Credit: Richard Houghton)

But one day, out of the blue, one of the project leaders dropped me an email with photos and said, Look, heres your mini brain. And there it was ... I wish there was some drama to it, but the researchers were pretty casual about it because they grow these things all the time. And I wish I could say that I remember thinking, Oh my God, this is my second brain, but I think I was more relieved than anything that it actually grew into an organoid. The fact that they actually got one that had these really clear, discernible structures in it was pretty remarkable. If anything, I felt a little bit proud of myself that we actually created something in the end.

For me, one of the really interesting things I gleaned from the project was that the stuff we are made from is incredibly versatile. The technology of transforming cells from virtually any other tissue in the body is relatively new, and its leading to all sorts of directions in medicine and research. I was fascinated to see firsthand what extraordinary things cells even mature, adult cells are capable of.

Read more here:
What It's Like to Grow a 'Mini-Brain' From Your Own Cells - Discover Magazine

Newborn in Japan receives first treatment with liver STEM cells – ZME Science

A team of doctors in Japan have successfully transplanted stem liver cells into a newborn baby who required transplant, marking a world first.

This approach could be used in the future for other infants who require organ transplants but are still too young or frail to bear such an intervention, the team explains. The patient suffered from urea cycle disorder, a condition where the liver is not able to break down ammonia, a toxic compound, in the blood, but was considered too small to survive a surgical intervention.

The success of this trial demonstrates safety in the worlds first clinical trial using human ES (embryonic stem) cells for patients with liver disease, said a press release of Japans National Center for Child Health and Development (NCCHD) following the procedure according to todayonline.

At only six days old, the infant (whose sex has hot been disclosed) was too small to undergo a liver transplant, which is not considered safe for patients under 6 kilograms (13 pounds), according to the NCCHD, which usually means they have to be around three to five months old.

However, the babys condition would have been fatal until then, so the doctors had to find an alternative way of treatment.

They settled on a bridge treatment meant to manage the condition until the baby was big enough for transplant. This procedure involved injecting 190 million liver cells derived from embryonic stem cells into the blood vessels of the liver. And it worked.

They report that the baby did not see an increase in blood ammonia concentrations after the procedure and grew up to successfully complete the next treatment, namely a liver transplant from its father. The patient was discharged from the hospital six months after birth.

This course of treatment can be used for infant (and perhaps adult) patients who are also waiting for a transplant in other parts of the world. Doctors at the NCCHD note that Europe and the US have a relatively stable supply of liver cells from brain-dead donors, while Japan only has a limited quantity to work with. So they had to use ES cells, which are harvested from fertilized eggs, which has caused some controversy regarding how ethical their use is.

The NCCHD is one of only two organisations in Japan allowed to work with ES cells to develop new medical treatments. It works with fertilised eggs whose use has been approved by both donors having already completed fertility treatment, according to the institute.

The treatment so far isnt meant to replace transplants, but thats definitely an exciting possibility for the future. Transplants save lives, but they rely on donors (whose numbers are limited) and require highly specialized equipment, doctors, and medicine to be successful. We can, however, hope that in the future a simple injection may replace the transplants of today.

Original post:
Newborn in Japan receives first treatment with liver STEM cells - ZME Science

Adult Stem Cells Market with Coronavirus (Covid-19) Impact Analysis | Industry Strong Development By Major Eminent Players, New Innovations, Key…

Adult Stem Cells Market 2020 this report is including with the COVID19 Outbreak Impact analysis of key points influencing the growth of the market. Also, report providing market data derived from primary as well as secondary research techniques. The report aims to deliver premium insights, quality data figures and information in relevance with aspects such as market scope, size, share, segments including types of products and services, application, geographies as well. It presents the 360-degree overview of the competitive landscape of the industries. SWOT analysis has been used to understand the strength, weaknesses, opportunities, and threats in front of the businesses. Thus, helping the companies to understand the threats and challenges in front of the businesses. Adult Stem Cells market is showing steady growth and CAGR is expected to improve during the forecast period.

This Adult Stem Cells Market Report That Is Imagines That the Length of This Market Will Develop during The Time System While the Compound Annual Growth Rate (CAGR) Development. The Adult Stem Cells Business Report Point Would Be the Economic Situations and Relating Orders and Takes the Market Players in Driving Fields Over the World.

The Major Players in the Adult Stem Cells Market.GlobalstemJuventas Therapeutics Inc.Epistem Ltd.Hybrid Organ GmbhCellerix SaMesoblast Ltd.Intellicell Biosciences Inc.NeuralstemCelyadCapricor Inc.ClontechCellerant Therapeutics Inc.Cellular Dynamics InternationalBiotime Inc.Beike Biotechnology Co. Ltd.Brainstorm Cell Therapeutics Inc.NeurogenerationInternational Stem Cell Corp.Gamida Cell Ltd.Caladrius Biosciences Inc.Cytori Therapeutics Inc.

Key Businesses Segmentation of Adult Stem Cells Market

Most important types of Adult Stem Cells products covered in this report are:Epithelial stem cellsHematopoietic stem cells

Most widely used downstream fields of Adult Stem Cells market covered in this report are:Neurodegenerative diseasesHeart diseaseBone diseaseOthers

Which prime data figures are included in the Adult Stem Cells market report?

What are the crucial aspects incorporated in the Adult Stem Cells market report?

Who all can be benefitted out of this Adult Stem Cells market report?

Research Goals:

The Report on Global Adult Stem Cells Market Studies the Strategy Pattern Adopted by Prominent International Players. Additionally, The Report Also Evaluates the Market Size in Terms of Revenue (USD MN) For the Forecast Period. All Data and Figures Involving Percentage Shares Splits, And Breakdowns Are Determined Using Secondary Sources and Verified Through Primary Sources.

Purchase FULL Report Now! https://www.qurateresearch.com/report/buy/HnM/global-adult-stem-cells-industry/QBI-MR-HnM-517105

A free report data (as a form of Excel Datasheet) will also be provided upon request along with a new purchase.

Contact Us:Web: http://www.qurateresearch.comE-mail: [emailprotected]Ph: US +13393375221, IN +919881074592

Go here to read the rest:
Adult Stem Cells Market with Coronavirus (Covid-19) Impact Analysis | Industry Strong Development By Major Eminent Players, New Innovations, Key...

Stem Cell and Regenerative Therapy Market Covid-19 Impact Analysis, Size, Share & Trends Analysis Report by Component, By Enterprise Size, By End…

ReportsnReports recently added a detailed overview and industry professional survey report on the global Stem Cell and Regenerative Therapy Market. In this report, titled Stem Cell and Regenerative Therapy Market Size, Share and Industry Analysis by Technologies, By Product, By Application, By Distribution Channel, and Regional Forecast 2019-2026.

Download a Sample Copy of Report athttps://www.reportsnreports.com/contacts/requestsample.aspx?name=2351731

The scope of the report encompasses the major types of Stem Cell and Regenerative Therapy Market that have been used, as well as the major applications being developed by industry, academic researchers and their commercialization offices, and government agencies. It analyzes the current market status, examines future market drivers, and presents forecasts of growth over the next five years. Technology developments, including the latest trends, are discussed. Other influential factors such as screening strategies for pharmaceuticals have also been included.

The global Stem Cell and Regenerative Therapy Market is comprehensively profiled in the report, including a detailed study of the markets key drivers and restraints, major market players, and leading segments.

Report Scope:

The scope of this report is broad and covers various type of product available in the stem cell and regenerative medicines market and potential application sectors across various industries. The current report offers a detailed analysis of the stem cell and regenerative medicines market.

The report highlights the current and future market potential of stem cell and regenerative medicines and provides a detailed analysis of the competitive environment, recent development, merger and acquisition, drivers, restraints, and technology background in the market. The report also covers market projections through 2024.

The report details market shares of stem cell and regenerative medicines based on products, application, and geography. Based on product the market is segmented into therapeutic products, cell banking, tools and reagents. The therapeutics products segments include cell therapy, tissue engineering and gene therapy. By application, the market is segmented into oncology, cardiovascular disorders, dermatology, orthopedic applications, central nervous system disorders, diabetes, others

The market is segmented by geography into the following regions: North America, Europe, Asia-Pacific, South America, and the Middle East and Africa. The report presents detailed analyses of major countries such as the U.S., Canada, Mexico, Germany, the U.K. France, Japan, China and India. For market estimates, data is provided for 2018 as the base year, with forecasts for 2019 through 2024. Estimated values are based on product manufacturers total revenues. Projected and forecasted revenue values are in constant U.S. dollars, unadjusted for inflation.

Report Includes:

28 data tables An overview of global markets for stem cell and regenerative medicines Analyses of global market trends, with data from 2018, estimates for 2019, and projections of compound annual growth rates (CAGRs) through 2024 Details of historic background and description of embryonic and adult stem cells Information on stem cell banking and stem cell research A look at the growing research & development activities in regenerative medicine Coverage of ethical issues in stem cell research & regulatory constraints on biopharmaceuticals Comprehensive company profiles of key players in the market, including Aldagen Inc., Caladrius Biosciences Inc., Daiichi Sankyo Co. Ltd., Gamida Cell Ltd. and Novartis AG

Any Query or Discount? Ask our Expert athttps://www.reportsnreports.com/contacts/discount.aspx?name=2351731

Summary:

The global market for stem cell and regenerative medicines was valued at REDACTED billion in 2018. The market is expected to grow at a compound annual growth rate (CAGR) of REDACTED to reach approximately REDACTED billion by 2024. Growth of the global market is attributed to the factors such as growingprevalence of cancer, technological advancement in product, growing adoption of novel therapeuticssuch as cell therapy, gene therapy in treatment of chronic diseases and increasing investment fromprivate players in cell-based therapies.

In the global market, North America held the highest market share in 2018. The Asia-Pacific region is anticipated to grow at the highest CAGR during the forecast period. The growing government funding for regenerative medicines in research institutes along with the growing number of clinical trials based on cell-based therapy and investment in R&D activities is expected to supplement the growth of the stem cell and regenerative market in Asia-Pacific region during the forecast period.

Reasons for Doing This Study

Global stem cell and regenerative medicines market comprises of various products for novel therapeutics that are adopted across various applications. New advancement and product launches have influenced the stem cell and regenerative medicines market and it is expected to grow in the near future. The biopharmaceutical companies are investing significantly in cell-based therapeutics. The government organizations are funding research and development activities related to stem cell research. These factors are impacting the stem cell and regenerative medicines market positively and augmenting the demand of stem cell and regenerative therapy among different application segments. The market is impacted through adoption of stem cell therapy. The key players in the market are investing in development of innovative products. The stem cell therapy market is likely to grow during the forecast period owing to growing investment from private companies, increasing in regulatory approval of stem cell-based therapeutics for treatment of chronic diseases and growth in commercial applications of regenerative medicine.

Products based on stem cells do not yet form an established market, but unlike some other potential applications of bioscience, stem cell technology has already produced many significant products in important therapeutic areas. The potential scope of the stem cell market is now becoming clear, and it is appropriate to review the technology, see its current practical applications, evaluate the participating companies and look to its future.

The report provides the reader with a background on stem cell and regenerative therapy, analyzes the current factors influencing the market, provides decision-makers the tools that inform decisions about expansion and penetration in this market.

Click Here & Get a Single User OR Corporate User License Key for This Report athttps://www.reportsnreports.com/purchase.aspx?name=2351731

The latest Stem Cell and Regenerative Therapy Market report provides readers with a deeper understanding of potential target consumers to create a lucrative marketing strategy for the 2019-2026 forecast period. For entrepreneurs seeking information about potential customers, it will be particularly helpful. Selective statements provided by leading vendors would allow entrepreneurs to gain a deeper understanding of the local market and prospective customers.

Table of Contents:

Chapter 1 Introduction

Study Background

Study Goals and Objectives

Reasons for Doing This Study

Scope of Report

Methodology and Information Sources

Geographic Breakdown

Market Breakdown

Analysts Credentials

.Continued

About Us: ReportsnReports.com is your single source for all market research needs. Our database includes 500,000+ market research reports from over 95 leading global publishers & in-depth market research studies of over 5000 micro markets.We provide 24/7 online and offline support to our customers.

E-mail: [emailprotected]

Phone: +1 888 391 5441

Visit link:
Stem Cell and Regenerative Therapy Market Covid-19 Impact Analysis, Size, Share & Trends Analysis Report by Component, By Enterprise Size, By End...

The RNA binding protein CPEB2 regulates hormone sensing in mammary gland development and luminal breast cancer – Science Advances

INTRODUCTION

The mammary gland develops postnatally and is subjected to marked remodeling in every oestrus cycle and during pregnancy. The mature mammary duct consists of an outer layer of basal myoepithelial cells and a polarized inner layer of luminal epithelial cells, which surround a hollow lumen and include hormone-sensing cells. During lactation, the lobuloalveolar units contain the luminal milk-producing alveolar cells (1, 2). This epithelial ductal tree is embedded within the mammary fat pad, which comprises fibroblasts, adipocytes, blood vessels, nerves, and immune cells (1). The development and remodeling of mammary ducts, through ductal branching and elongation, require epithelial cell proliferation to be coordinated with specification and maintenance of cell differentiation, as well as with tissue and cell polarity. These events are governed by ovarian steroid hormones, which control normal mammary development and lead to the neoplastic conversion of mammary tissue when misregulated. Estrogen is the most potent mitogenic stimulus for mammary ductal elongation during puberty, and it also directs the transcription of progesterone receptor (PR), which, in turn, induces ductal side branching and luminal lineage differentiation (35). Hormone-sensing cells, which are positive for estrogen receptor (ER) and PR, account for only a small fraction (7 to 30%) of the luminal epithelium. These hormone receptorpositive (HR+) cells integrate hormonal cues to signal to adjacent HR-negative (HR) cells via paracrine communication, which trigger the major proliferative response at the adult stage, mainly through the receptor activator of nuclear factor B (NFB) ligand (RANKL) (68).

Temporal and spatial control of mRNA translation, coupled to regulation of mRNA stability and localization, link cell proliferation, polarity, and differentiation (912). These gene regulation responses and the integration of external signals are coordinated through RNA binding proteins and cognate cis-acting elements to assemble specific ribonucleoprotein complexes. The cytoplasmic polyadenylation element (CPE)binding (CPEB) family of RNA binding proteins regulates mRNA stability and translation through dynamic changes in their poly(A) tail length (13, 14). The four family members (CPEB1 to CPEB4) competitively recognize the same CPE in the 3 untranslated region (3UTR) of target mRNAs (15). CPEs interact with other cis-elements in a CPE combinatorial code to define spatiotemporal gene expression patterns (11, 1619). In turn, individual pairs of CPE/CPEBs assemble into complexes that either repress or activate translation; repressor complexes shorten the poly(A) tail and mediate subcellular localization of repressed mRNAs, while activator complexes elongate the poly(A) tail (13). The switch from repression to activation is regulated by coordinated CPEB-specific posttranslational modifications of all four CPEBs (20). Although most CPEB functions have been studied during early development, CPEB1 in the mammary gland regulates the translation of milk protein transcripts, such as -casein mRNA (21), and the localizationbut not the translational activationof ZO-1 (Zona Occludens Protein 1) mRNA to the apical surface of epithelial cells for tight junction assembly (22). Changes in poly(A) tail length regulate gene expression, integrating extracellular signals into cellular outcomes, including mitotic cell division and steroid hormone responses (17, 23, 24). Here, we show that the RNA binding protein CPEB2, which regulates the poly(A) tail length of CPE-containing mRNAs, contributes to mammary gland development and luminal breast carcinogenesis by regulating the translation of mRNAs downstream of steroid hormone signaling.

To address how CPEBs could contribute to postnatal mammary gland development, we first determined the relative expression levels of all four CPEB mRNAs in pubertal, adult, pregnant, lactating, and involuted mouse mammary glands (Fig. 1A). Cpeb2 mRNA was the most abundant of the four Cpeb mRNAs in adult virgin mice, and it also peaked at lactation. After cell sorting of mammary epithelial cells (MECs) (fig. S1A), we found that Cpeb2 mRNA was expressed mainly in luminal cells, whereas Cpeb1 was predominant in myoepithelial cells (Fig. 1B). A similar distribution was observed at the protein level (fig. S1B). We next determined the consequences in mammary gland morphogenesis of total loss-of-function mouse models for CPEBs in postpubertal adult nulliparous mice. To this end, we determined the elongation and branching of the epithelial ductal tree in mammary gland whole mounts. We used previously described knockout (KO) mice for CPEB1 and CPEB4 (19, 25) and generated KO mice for CPEB2 and CPEB3 (figs. S2 and S3). CPEB2 and CPEB3 KO mice were viable and fertile and did not show any overt phenotype. While ductal morphogenesis was not affected in CPEB3 KO or CPEB4 KO mice, CPEB1 KO and CPEB2 KO animals displayed reduced branching through the fat pad (Fig. 1C and fig. S4A). Branching was quantified using AngioTool software (fig. S4B). Because of a defect in oogenesis, ovaries from CPEB1 KO females are rudimentary and do not secrete normal levels of reproductive hormones (26). This deficiency, which can be partially rescued by injection of 17-estradiol (22), limits mammary duct proliferation. Accordingly, we observed reduced ductal expansion through the fat pad only in adult CPEB1-deficient mammary glands (fig. S4). To better define cell-autonomous defects in mammary duct development, we generated CK14-specific KO mice for CPEB1 and CPEB2 (KOCK14), where the CK14 promoter is expressed by all MECs during embryonic development (27). When the KO was restricted to the CK14 lineage, loss of CPEB2 (but not of CPEB1) resulted in reduced number of junctions (Fig. 1D). At earlier developmental times, we also observed a delayed ductal expansion in CPEB2 KO mice, as shown by diminished pubertal invasion of the epithelial tree through the fat pad that was recovered in adulthood (Fig. 1E and fig. S4B). CPEB2 KO mice also showed an increased luminal/myoepithelial cell ratio (Fig. 1F and fig. S4D). Thus, deletion of CPEB2 results in delayed ductal extension and reduced branching, two events sequentially regulated by ER and PR.

(A) mRNA levels of Cpeb1 to Cpeb4 normalized to Gapdh in whole tissue mammary gland (n = 2; n = 7 for adult nulliparous). Tissue was obtained from mice at puberty (5 weeks old), adult nulliparous (10 weeks old), midpregnancy (day 12 of gestation), lactation (2 weeks of lactation), or involution (6 days after weaning). Gapdh expression is also shown. Statistics were determined using two-way analysis of variance (ANOVA), **P < 0.01, ***P < 0.001, and ****P < 0.0001. (B) mRNA levels of Cpeb1 to Cpeb4 normalized to Gapdh in sorted cells from adult virgin mammary gland (n = 3). Statistics using two-way ANOVA, ****P < 0.0001. Myo, myoepithelial. (C) Representative carmine-stained mammary gland whole mounts and automatic quantification of the number junctions in virgin 10- to 12-week-old WT (n = 11) and constitutive CPEB1 KO (n = 4), CPEB2 KO (n = 10), CPEB3 KO (n = 5), and CPEB4 KO (n = 4) mice. Statistics were determined using the Mann-Whitney test, *P < 0.05 and **P < 0.01. (D) Representative mammary whole mounts and automatic quantification of the number of junctions in virgin 10- to 12-week-old epithelial-specific WTCK14 (n = 4), CPEB1 KOCK14 (n = 6), and CPEB2 KO CK14 (n = 8) mice. Statistics were determined using the Mann-Whitney test, *P < 0.05. (E) Representative mammary whole mounts and quantification of the area of the fat pad filled with epithelial ducts at puberty in WT and CPEB2 KO females (5 weeks old) (n = 5). Statistics were determined using the Mann-Whitney test, *P < 0.05. (F) Ratio between the percentage of luminal and myoepithelial cells gated on lineage-negative (WT, n = 7; CPEB1 KO, n = 4; CPEB2 KO, n = 6; CPEB3 KO, n = 4; and CPEB4 KO, n = 4). Statistics were determined using the Mann-Whitney test, *P < 0.05.

To further determine the cell-of-origin of the mammary CPEB2 KO phenotype and given that CPEB2 was mostly expressed in the luminal compartment of the mammary gland (Fig. 1B and fig. S1B), we sorted luminal cell types from adult virgin mammary glands (28). We distinguished the following three cell types [as defined in (28, 29)]: ductal progenitor (DP; Sca1+CD49b+), ductal differentiated (DD; Sca1+CD49b), and alveolar progenitor (AP; Sca1CD49b) (Fig. 2A). We observed a general increase in Sca1 levels in CPEB2 KO mammary glands and increased cell number in the gate for the Sca1+CD49b+ population, concomitant with a reduction in the Sca1+CD49b window (Fig. 2, A to E). The AP population, on the other hand, did not change significantly upon CPEB2 depletion. To further characterize the effect of CPEB2 loss-of-function in MECs, we studied the transcriptomes of all four wild-type (WT) and CPEB2 KO epithelial populations using DNA microarrays. First, we confirmed our gating strategy through the expression of well-known markers in the expected populations (fig. S5A). Principal components analysis of gene expression profiles further confirmed clustering by populations and showed that the main differences between WT and CPEB2 KO cells affected the Sca1+CD49b+ population, with DPKO placed between DPWT and DDWT (fig. S5B). This was calculated by comparing the distance between centroids of different genotypes on a given population versus the dispersion within the population (see Methods and fig. S5B). Next, on the basis of the genes differentially expressed in the DPWT versus DDWT populations, we generated a WT progenitor signature by selecting candidate genes with the highest and lowest fold change (FC) percentiles and P < 0.01 (1% most up- and down-regulated genes, n = 181 and n = 101, respectively). We found a clear negative enrichment for the WT progenitor signature in DPKO cells, with the genes up-regulated being negatively enriched and vice versa (Fig. 2F nd fig. S5C). Similarly, further filtering using a false discovery rate (FDR) of 0.1 as a threshold (instead of P value) resulted in a more stringent signature with 24 up-regulated and no down-regulated genes (WT DP versus WT DD) that was also negatively enriched in DPKO cells (fig. S5D) (see Methods). These observations suggest that the DPKO cells contained a partially differentiated population. mRNA expression of the luminal progenitor markers Elf5, Kit, Cd14, and Rspo1 (29) was reduced in DPKO cells as compared with the DPWT population (Fig. 2G). Conversely, these luminal progenitor markers were unaffected in APKO cells, with the exception of Rspo1 (see Discussion) (Fig. 2H). Accordingly, DPKO cells showed a reduced capacity to form organoids as compared to DPWT cells (Fig. 2I). Together, these results indicated that CPEB2 might be required for the proper differentiation of DP cells.

(A) Representative fluorescence-activated cell sorting (FACS) plots gated on luminal cells depicting luminal subpopulations: ductal differentiated (DD; Sca1+CD49b), ductal progenitors (DPs; Sca1+CD49b+), and alveolar progenitors (APs; Sca1CD49b+). (B) Representative FACS plots for Sca1 gated on luminal cells. FSC-W, forward scatter width. (C) Ratio of the percentage of Sca1high and Sca1low populations in luminal cells (n = 10). Statistics were determined using the Mann-Whitney test, **P < 0.01. (D) Quantification of the percentage of CD49b+ cells gated on luminal cells (n = 17). Statistics were determined using the Mann-Whitney test, *P < 0.05. (E) Quantification of the luminal subpopulations as in (A). Statistics were determined using two-way ANOVA, ***P < 0.001 (n = 17). (F) Preranked GSEA graphical output for the enrichment in DPKO versus DPWT cells of the up-regulated genes in the WT progenitor signature generated by P value (n = 181, see Methods). FDR q 0.0001. FDR, false discovery rate; NES, normalized enrichment score. (G) Expression of luminal progenitor markers in DPWT and DPKO cells. (H) Expression of luminal progenitor markers in APWT and APKO cells. (I) Representative images of organoids from sorted DPWT and DPKO cells and automatic quantification of the number of organoids from sorted DD or DP cells. Scale bars, 100 m. Statistics were determined using two-way ANOVA, **P < 0.01.

Gene set enrichment analysis (GSEA) showed a clear down-regulation in the gene sets related to cell cycle and proliferation (G2M checkpoint and E2F targets) in all four CPEB2 KO epithelial cell populations (fig. S6A). DD cells are highly proliferative (30). Therefore, we next analyzed MEC proliferation in the CPEB2 KO by Ki67 immunostaining (Fig. 3A) and by 5-ethynyl-2-deoxyuridine (EdU) incorporation (Fig. 3B). CPEB2 KO mice displayed reduced MEC proliferation. Note that apoptosis was negligible in adult mammary glands, both in WT and CPEB2 KO animals (fig. S6B).

(A) Representative images and automatic quantification of Ki67+ cells by immunohistochemistry in adult virgin mammary gland in WT and CPEB2 KO (n = 7) mice. Statistics were determined using the Mann-Whitney test, *P < 0.05. Scale bars, 50 m. (B) Representative FACS plots (gated on MECs) and quantification of percentage of EdU incorporation. FSC-A, FSC area. Statistics were determined using two-tailed unpaired Students t test, *P < 0.05. MECs, mammary epithelial cells. (C) Representative images and automatic quantification of ER+ cells by immunohistochemistry in adult virgin mammary gland in WT and CPEB2 KO (n = 5). Statistics were determined using the Mann-Whitney test, *P < 0.05. Scale bars, 25 m. (D) Representative images and automatic quantification of PR+ cells by immunohistochemistry in adult virgin mammary gland in WT and CPEB2 KO (n = 5). Statistics were determined using the Mann-Whitney test, *P < 0.05. Scale bars, 25 m. (E) Preranked GSEA graphical output for the enrichment in Sca1+KO cells (DPKO + DDKO) of the gene set estrogen response early from the Molecular Signatures Database Hallmarks collection (see Methods). FDR q = 0.0139. (F) Heat map representing the log2FC expression of hormone-driven genes in DPKO compared to DPWT. (G) Cpeb2 expression levels normalized by Gapdh in epithelial subpopulations.

Proliferation in the mammary gland is driven by the action of steroid hormones not only for HR+ but also for HR cells (including mammary stem cells) through dominant paracrine effects (4, 31). Thus, we first assessed the levels of ER and PR in constitutive and CK14-driven CPEB2 KO mice. Unexpectedly, ER and PR were up-regulated in the absence of CPEB2, both at mRNA and protein levels (Fig. 3, C and D and fig. S6, C to E). Moreover, the hallmark estrogen response early was significantly increased in KO Sca1+ cells (Fig. 3E), suggesting that the ER transcriptional function was not impaired. Direct ER and PR target genes tended to be up-regulated in the absence of CPEB2 at the transcript levels, while downstream proliferative genes were down-regulated (Fig. 3F). These observations suggest that, although hormone-receptor transcriptional activity is normal, or even increased, the downstream effectors of hormone-driven cell proliferation are defective.

We found that, in the absence of CPEB2, there is a delay in ductal elongation at puberty, as well as reduced ductal branching in adulthood, accompanied by decreased epithelial proliferation and impaired differentiation of HR+ cells. All these phenotypes observed in vivo are concordant with blunted HR signaling (4). Given that CPEB2 was expressed mainly in HR+ cells (Fig. 3G and fig. S7A), we hypothesized that CPEB2 may constitute a previously unidentified posttranscriptional layer of regulation in the ER and PR pathways.

To identify the CPEB2-target mRNAs that could explain the defective response to hormones in MECs, we performed CPEB2 RNA immunoprecipitation (RIP; Fig. 4A). CPEB2 coimmunoprecipitated 169 mRNAs in MECs, which were significantly enriched in the RIP WT compared with the RIP in CPEB2 KO control cells (see Methods, table S1, and fig. S7B). These CPEB2 targets were enriched in canonical CPEs (UUUUA12U), thereby verifying the specificity of the immunoprecipitation (Fig. 4B). Pathway analysis showed that CPEB2-target mRNAs were enriched in breast cancerrelated genes (Fzd2, Jag1, Cdk6, Ccnd1, Sp1, Wnt5a, Kit, Kras, and Lrp6) (Fig. 4C). RIP targets were also overrepresented in the phosphoinositide 3-kinase (PI3K)Akt signaling pathway (Fig. 4C), which has been shown to modulate both genomic and nongenomic activities of the ER and is associated with breast cancer and with endocrine resistance of luminal tumors when mutated (32). The transcription factor 3,5-cyclic adenosine monophosphate OR cyclic adenosine monophosphat responsive element binding protein 1 (CREB1), which is activated downstream PI3K-Akt and regulates estrogen signaling (33, 34), was one of the top three enriched transcripts in the RIP WT (table S1 and fig. S7B). Moreover, individual targets included not only Cpeb2 and Cpeb3 mRNAs (suggesting auto- and cross-feedback CPEB loops) but also regulators of cell fate, morphogenesis, and organogenesis in the Wnt and Notch pathways (1, 35), such as the Wnt surface receptors Fzd2 and Lrp6, and the Notch surface ligand Jag1 (table S1 and fig. S7B). Furthermore, although not statistically significant due to low mRNA expression levels, Rankl (Tnfsf11) was enriched in the CPEB2 RIP, and we also found CyclinD1 (Ccnd1) to be a CPEB2 target (fig. S7B). Rankl and Ccnd1 are the key effectors of the autocrine and paracrine proliferative responses to progesterone, respectively. We validated several of these genes as bona fide CPEB2 target mRNAs by RIPquantitative polymerase chain reaction (qPCR) (Fig. 4D). Given their direct implications on the regulation of hormone-driven proliferation and differentiation in MECs, we further analyzed the regulation of Creb1, Ccnd1, and Rankl. These CPEB2 target mRNAs contained conserved canonical CPEs in their 3UTRs at optimal distances (17) from the polyadenylation sites (fig. S8A). We found that their protein levels were reduced in the absence of CPEB2, without significant variations in their mRNA levels, thereby suggesting translational changes (Fig. 4, E to H and fig. S8B). This CPEB2-mediated regulation of RANKL appeared to be specific for MECs, given that it was not observed in the immune cells of the mammary lymph node (fig. S8C).

(A) Western blot image for CPEB2 and vinculin (as a control) from unbound, input, and immunoprecipitated fractions with anti-CPEB2 antibody in WT and CPEB2 KO MECs. (B) Percentage of genes with (+CPEs, red) or without (CPEs, gray) CPEs in the 3UTR, comparing RIP targets to the mouse transcriptome (all). Statistics were determined using Fishers exact test, ****P < 0.0001. (C) Significantly enriched KEGG pathways (adjusted P < 0.05) in the analyzed RIP targets. cGMP-PKG, cyclic guanosine monophosphate (cGMP)cGMP-dependent Protein Kinase G (PKG). mTOR, mammalian target of rapamycin. (D) RIP-qPCR results showing the RIP values normalized by each input in WT (n = 4) and KO (n = 3) MECs. Gapdh mRNA and RIP in CPEB2 KO MECs are used as negative controls for enrichment in RIP as compared to input. Statistics were determined using the Mann-Whitney test, *P < 0.05. IP, immunoprecipitation. (E) Western blot image for CPEB2, CREB1, and -tubulin (loading control) and normalized quantification of CREB1 protein levels in WT and KO MECs (n = 6). Statistics were determined using the Mann-Whitney test, **P < 0.01. (F) Representative images and manual quantification of RANKL+ cells by immunohistochemistry in adult virgin mammary gland in WT and CPEB2 KO animals (n = 6). Scale bar, 50 m. Statistics were determined using the Mann-Whitney test, **P < 0.01. (G) Western blot image for CPEB2, CyclinD1, and -tubulin (loading control) and normalized quantification of CyclinD1 protein levels in WT and KO MECs (n = 6). Statistics were determined using the Mann-Whitney test, *P < 0.05. (H) mRNA levels of Rankl, Ccnd1, and Creb1 normalized to Gapdh and to WT in MECs (WT, n = 6; KO, n = 4). Statistics were determined using the Mann-Whitney test.

As CPEB2 KO mice displayed defective signaling to estrogen and progesterone, both key in breast cancer development (29, 36, 37), and CPEB2-bound mRNAS were components of breast cancer pathways, we next explored whether CPEB2 participates in breast tumorigenesis. Analysis of the expression of CPEB2 mRNA in patient breast tumor samples using the METABRIC cohort determined an association between CPEB2 and ESR1 levels (Fig. 5A). In agreement with the function of CPEB2 in mammary homeostasis, gene expression profiles that classify breast cancer into various subtypes (38) indicate that ER+ primary breast cancer has a characteristic luminal transcriptional profile. Using both the METABRIC and The Cancer Genome Atlas RNA sequencing (RNA-seq) dataset, we confirmed that CPEB2 levels were decreased in basal-like and Her2 tumors compared to luminal tumors and to morphologically normal surrounding tissue (Fig. 5B and fig. S9A). This observation was extended to human breast cancer cell lines, with several ER+ (luminal-like) cell lines expressing higher levels of CPEB2 mRNA (Fig 5C).

(A) Violin plots for CPEB2 RNA expression depending on ER status; METABRIC cohort (n = 1974). Statistics were determined using the Wald test, P < 10 2.2216. (B) Violin plots for CPEB2 RNA expression in the PAM50 subtypes; METABRIC cohort (n = 1974). Statistics were determined using the Wald test compared to the luminal A subtype: basal-like, P < 10 2.2216; HER2, P < 10 2.2216; and luminal B, P = 0.99003. (C) Quantification of CPEB2 expression levels by RT-qPCR in the indicated breast cancer cell lines. B2M was used as endogenous control. (D) Kaplan-Meier survival curves for patients with luminal A breast cancer [HR (<10 years) = 1.89; P = 0.021; multivariate using tumor size and lymph node as other risk factors n = 550]. (E) Schematic representation of the chemical-induced breast cancer model and kinetics of mammary tumor onset in mice treated with medroxyprogesterone acetate (MPA) and 7,12-dimethylbenz(a)anthracene (DMBA) as indicated. Statistics were determined using the log-rank test, *P < 0.05. (F) Number of macroscopic tumors per animal at time of sacrifice (16 weeks after MPA administration) in WTCK14 (n = 11) and CPEB2 KOCK14 (n = 11) animals. Statistics were determined using the Mann-Whitney test, *P < 0.05. (G) Tumor incidence in WTCK14 (n = 11) and CPEB2 KOCK14 (n = 11) mice. Statistics were determined using chi-square test, *P < 0.05. (H) Western blot image for CPEB2 and vinculin (loading control) in ZR75 cells after KD of CPEB2 using sh_CPEB2 #28 or #78 or in control cells (sh_Control). (I) Relative growth curve of ZR75 cells sh_Control or KD of CPEB2. Cell numbers were quantified relative to day 0 at the indicated time points. Statistics were determined using a two-tailed unpaired Students t test, ***P < 0.001. (J) Surviving fraction of CPEB2 KD ZR75 cells (using sh_CPEB2 #28 and #78) or control ZR75 cells treated with vehicle (0 M), 0.5 M 4-OHT, or 1 M 4-OHT. Number of viable cells was quantified 6 days after 4-OHT treatment. Surviving fraction refers to the fraction of cells present after 4-OHT treatment. Statistics were determined using a two-tailed unpaired Students t test, *P < 0.05 and ***P < 0.001. n.s., not significant. (K) RT-qPCR quantification of MYC expression levels in CPEB2 KD ZR75 cells (sh_CPEB2 #28 or #78) or control ZR75 cells (sh_Control) treated with vehicle (0 M) or 1 M 4-OHT for 48 hours. B2M was used as an endogenous control. Statistics were determined using a two-tailed unpaired Students t test, **P < 0.01 and ***P < 0.001. (L) Quantification of CCND1 expression levels by RT-qPCR in CPEB2 KD ZR75 cells (sh_CPEB2 #28 and #78) or control cells (sh_Control) treated with vehicle (0 M) or 1 M 4-OHT for 48 hours. B2M was used as an endogenous control. Statistics were determined using a two-tailed unpaired Students t test, *P < 0.05, **P < 0.01, and ***P < 0.001.

Next, we explored the association between CPEB2 expression and patient survival at 10 years using the METABRIC public breast cancer primary tumor cohort, for which prognosis annotation was available with sufficient follow-up. We confirmed an interaction between CPEB2 expression and samples classified on the basis of PAM50 molecular subtype (P = 0.0007, continuous model) (39), implying significant differences in prognosis association across biologically diverse tumor subtypes. In luminal A tumors, dependent on ER signaling for growth, high levels of CPEB2 were associated with worse survival compared to samples with the lowest expression [HR (<10 years) = 1.83, P = 0.028, n = 550; Fig. 5D]. No association between CPEB2 expression and tumor size was observed (fig. S9B). Collectively, these findings reveal an association between low CPEB2 expression and survival in patients with luminal ER+ breast cancer.

To experimentally address a potential role of CPEB2 in luminal tumorigenesis, we induced mammary tumor development in WTCK14 and CPEB2 KOCK14 mice, combining the proliferative action of the synthetic progestin medroxyprogesterone acetate (MPA) and the mutagenic agent 7,12-dimethylbenz(a)anthracene (DMBA) (40). Tumor onset was significantly delayed in CPEB2 KOCK14 mice (Fig. 5E), as shown by the higher percentage of tumor-free animals at 20 weeks after MPA treatment, the humane end point determined by the size of WT tumors. Tumor incidence was 63% for WTCK14 animals versus 27% for CPEB2CK14 mice. Moreover, at the end of the experiment, the number of tumors per animal (Fig. 5F) was reduced CPEB2CK14 animals. As previously described (41), these treatments generated hyperplasias, neoplasias, adenomas, adenocarcinomas, and adenosquamous carcinomas. Histopathological analysis of the tumors generated in the CPEB2 KOCK14 and WTCK14 animals revealed no major differences (fig. S9C). Furthermore, we detected lower ER levels in CPEB2 KOCK14 tumors as compared to the WTCK14 ones (fig. S9D), despite the fact that this treatment generates tumors characteristic of the luminal breast cancer subtype with high ER expression (42) (note that determination of significance was limited due to low number of tumors in the CPEB2 KOCK14 mice).

To further explore any functional interactions between ER and CPEB2, we knocked down CPEB2 in ZR75 ER+ luminal human breast cancer cells using two independent short hairpin RNAs (shRNAs; Fig. 5H and fig. S10A). These depletions significantly decreased cell proliferation in vitro but did not increase apoptosis (Fig. 5I and fig. S10B). Next, we treated WT and CPEB2 knockdown (KD) cells with the ER inhibitor 4-hydroxytamoxifen (4-OHT) (Fig. 5J). In contrast to WT ZR75 cells, CPEB2 KD ZR75 cells were insensitive to 4-OHT, thereby indicating that CPEB2 depletion and ER signaling inhibition do not have an additive effect on cell growth and suggesting that CPEB2 and ER act on the same pathway. Consistently, the effects of CPEB2 depletion on MYC and CCND1 expression (genes regulated by ER signaling and mediators of proliferation) were comparable, but not additive, to inhibition of ER signaling by 4-OHT (Fig. 5, K and L). Furthermore, we could also validate the regulation of RANKL by CPEB2 in this breast cancer setting (fig. S10, C and D).

Our results indicate that CPEB2 and ESR1 expression in breast cancer are linked and that high CPEB2 levels are associated with poor prognosis in luminal A tumors. Results of MPA/DMBA tumor generation indicated that high CPEB2 expression promotes luminal tumor development, consistent with the hormone dependence of this breast tumor subtype. On the other hand, ER tumors (such as basal like) do not seem to require CPEB2; low levels of CPEB2 result in reduced survival (fig. S9E).

In this work, we unveil a previously unknown layer of posttranscriptional regulation of gene expression orchestrated by CPEB2 in the mammary epithelia hormone responses. Thus, key HR-driven mediators (both cell autonomous and paracrine) of the differentiation and proliferation pathways (such as RANKL, CyclinD1, or CREB1) are encoded by CPEB2-regulated mRNAs. In the absence of CPEB2, the transcriptional activation of these genes fails to be reflected into increased protein levels. Mammary ductal branching and elongation are coordinated by the ovarian steroid hormones estrogen and progesterone, which activate transcriptional programs resulting in epithelial cell differentiation and proliferation. These hormones are sensed by a minority of HR+ cells, which, in turn, signal to adjacent HR cells through paracrine signals that coordinate mammary gland development and remodeling. Although CPEB2 can modulate the expression of more than a hundred genes (table S1) rather than switching on a single gene, the depletion of this RNA binding protein shows phenotypic similarities with the depletion of well-characterized HR-activated genes. CyclinD1 and CREB1 determine the proliferative programs of the estrogen signaling in the mammary gland (34, 43). In turn, RANKL is a key paracrine mediator of progesterone-mediated ductal side branching and MEC proliferation (mediated by NFB and CyclinD1) and differentiation (6, 7, 44, 45). All of these pathways are defective in the absence of CPEB2. In addition to being a CPEB2 target in luminal cells, Ccnd1 is also down-regulated in myoepithelial cells, probably as the result of a paracrine transcriptional effect (fig. S11A). Expression of Rspo1, which was down-regulated in both DPKO and APKO (Fig. 2, G and H), is a RANKL-induced gene (6). Thus, the mammary epithelia defects observed in CPEB2 KO mice could be partly explained by impaired translational activation of Rankl mRNA. However, note that the phenotype of CPEB2 KO mouse model does not phenocopy that of the RANKL KO. RANKL drives mammary alveologenesis (46), which is not defective in CPEB2 KO mice (fig. S11, B and C). Normal alveologenesis in CPEB2 KO mice could be due to a compensatory increase in Cpeb4 mRNA levels, which we observed specifically at the lactating stage but not in adult virgin mammary glands (fig. S11D). Redundancy between CPEB2 and CPEB4 has been reported in other scenarios (47).

In this study, we have focused on the role of CPEB2 in luminal breast cancer as a mediator of ER signaling. Accordingly, CPEB2 is one of the top six genes, together with ESR1, with strongest correlation with ER+ breast cancer prognosis (48). It has been proposed that breast cancer subtypes arise from distinct epithelial differentiation stages and lineages (29). Although the cell-of-origin for luminal tumors has not yet been unambiguously identified, these tumors appear to arise from a population of DPs that not only has clonogenic capacity but also expresses high levels of markers of mature luminal cells, such as ER, PR, GATA3 (GATA binding protein 3), and FOXA1 (Forkhead Box Protein A1) (28, 29, 49, 50). Depletion of CPEB2 generated a differentiation intermediate population with high Sca1/ER levels but low clonogenic capacity and impaired hormonal signaling. Together, our findings reveal a previously unkown posttranscriptional mechanism that regulates mammary gland morphodynamics and influences the outcome of ER+ mammary tumors, which account for 75% of breast cancer cases.

To generate a CPEB2 KO mouse model, the vector (EUCOMM, PRPGS00036-W-3-B04) was electroporated in mouse G4 embryonic stem cells (mixed C57BL/6J and 129/Sv). Positive recombinant embryonic stem cells were identified by Southern blotting, transfected in vitro with the FlpO recombinase to remove the geo-cassette, and microinjected into developing blastocysts. Resulting chimeric mice (Cpeb2 lox/lox) were crossed with C57BL6/J mice, and the mouse colony was maintained in a mixed background (70% C57BL/6J and 30% 129/Sv). To generate CPEB3 KO, mouse ES cells carrying a gene-trap lacZ cassette and a promotor-driven neomycin resistance gene in Cpeb3 intron 3 (clones HEPD0670_2_C02 and HEPD0670_2_G03, EUCOMM) were microinjected into developing blastocysts. Resulting chimeric mice were crossed with 129/Sv C57Bl/6J animals. To obtain a ubiquitous and constitutive depletion, Cpeb2lox/lox mice were crossed with mice expressing DNA recombinase Cre under control of the Sox2 promoter. Excision of exon 4 of Cpeb2 led to a frameshift in the mRNA, generating premature stop codons and resulting in animals that were KO for the CPEB2 protein. For the CPEB3 KO, the Neo cassette and exon 3 were further deleted by crossing Cpeb3loxfrt with transgenic mice expressing Cre under the control of the Sox2 promoter. The mouse colony was maintained in a mixed background (129/Sv C57Bl/6). Epithelial-specific CPEB1 and CPEB2 KO mice were obtained by crossing Cpeb1lox/lox or Cpeb2lox/lox animals with C57BL/6J transgenic mice expressing Cre under control of the Krt14 promoter. Routine genotyping was performed by PCR; primer sequences are listed in table S2.

Agarose gels were incubated under soft agitation with depurination solution (0.25 M HCl, 15 min), denaturation solution (1.5 M NaCl and 0.5 M NaOH, 45 min), and neutralization solution (0.5 M tris and 1.5 M NaCl, 30 min). After overnight transfer, DNA was cross-linked (254 nm, 0.12 J) to a nylon membrane (0.45 mm; Pall Corporation). The membrane was prehybridized with Church buffer for 3 hours at 65C, hybridized with 32P-labeled probes for 12 hours, rinsed with washing buffer (standard saline citrate, 0.1% SDS), and exposed to a phosphorimager screen.

Mice (Mus musculus, C57BL/6J-129/Sv mixed background) were maintained under a standard 12-hour light/12-hour dark cycle at 23C, with free access to food and water. Female littermates between 10 and 12 weeks of age were used, unless otherwise stated. Mice were staged by histological analysis of ovaries or vaginal cytology and were selected for the follicular phase of the oestrous cycle (51, 52). For tumorigenesis experiments, CK14-Creexpressing mice were subcutaneously injected with MPA (Depo-Provera) at 7 weeks of age. They were then given DMBA (1 mg) by gavage weekly during the following 4 weeks (53, 54). Tumors were detected and monitored by manual palpation. Mice were sacrificed when a palpable mass exceeded 1 cm in diameter or at 20 weeks after MPA treatment (time for many WT animals to develop tumors reaching this humane end point). End-point tumors were classified on the basis of previously identified pathological nomenclature (55).

Thoracic and inguinal mammary glands were dissected, and MECs were prepared as previously described (56). In brief, mammary glands were incubated with a collagenase/hyaluronidase solution (STEMCELL Technologies), red blood cells were lysed, and cells were further dissociated with trypsin (Sigma-Aldrich), dispase II (Sigma-Aldrich), and deoxyribonuclease I (Sigma-Aldrich). In general, fluorescence-activated cell sorting (FACS) analysis and sorting were performed in a FACS Aria Fusion sorter (BD Biosciences), and data were analyzed with the BD FACSDiva software. For four-color FACS analysis, a Gallios flow cytometer (Beckman Coulter) was used, and data were analyzed with the FlowJo software. The following antibodies were used: EpCAMphycoerythrin (PE) (130-102-265), CD49fallophycocyamin (APC) (130-100-147), CD45fluorescein isothiocyanate (FITC) (130-102-778), Ter119-FITC (130-102-257), CD31-FITC (130-102-970), CD49b-PE (130-102-778), EpCAM-APC/Cy7 (BioLegend, 118217), and Ly-6A/E (Sca1) PerCP/Cy5.5 (BioLegend, 108123). Antibodies were purchased from Miltenyi Biotec unless otherwise stated. Gating strategies were adjusted as previously described (28). For EdU incorporation experiments, mice received an intraperitoneal injection of EdU (80 mg kg1) and were sacrificed 6 hours later, as previously described (57). After isolation of MECs, samples were processed as indicated in the protocol for Click-iT Plus EdU Flow Cytometry Assay (Invitrogen) using Pacific Blue picolyl azide.

A total of 2000 sorted cells were embedded in one drop of basement membrane extracts (Cultrex) and cultured for 15 days in uncoated 24-well glass plates (no. 242-20, zell-kontakt). The culture protocol was adapted from (58); advanced Dulbeccos modified Eagle medium (DMEM)/F12 medium was supplemented with penicillin/streptomycin, GlutaMAX, Hepes (Gibco), hydrocortisone (Lonza Bioscience), B27 (Thermo Fisher Scientific), insulin, N-acetylcysteine, epidermal growth factor, fibroblast growth factor 2 (FGF2; Sigma-Aldrich), FGF10 (PeproTech), heparin (STEMCELL Technologies), Y-27632 (ROCK inhibitor, Tocris), Wnt3a, and R-spondin1 (in-house). ROCK inhibitor was added for the first week, and the medium was refreshed every 3 to 5 days. Full drops were scanned with an Olympus IX81 inverted microscope at 10 magnification (ScanR software). Bright-field Z stacks of each field were projected in a single image, and the full drop was then digitally reconstructed by stitching the different image projections using an ImageJ custom-made macro-developed for this purpose at the Institute of Research in Biomedicine (IRB) Advanced Digital Microscopy Facility.

For mammary gland whole mounts, inguinal mammary glands were placed on a slide and fixed immediately with Carnoys solution overnight. Tissue was then hydrated, stained with carmine alum (Sigma-Aldrich, C1022 and A7167), dehydrated, cleared with xylene, and mounted with Leica CV Mount (14046430011). Images from whole mounts were acquired with an Olympus macroscope (zoom 1.6) and joined with the MosaicJ tool from ImageJ (59). For junction quantification, images were processed using an ImageJ custom-made macro-developed for this purpose and then analyzed using AngioTool (60). For histology and immunohistochemistry, inguinal mammary glands were fixed in 10% neutral-buffered formalin solution and embedded in paraffin. Paraffin-embedded tissue sections (3 m in thickness) were first air-dried and then dried at 60C overnight. Immunohistochemistry was performed using Autostainer Plus (Dako, Agilent). Before immunohistochemistry, sections were dewaxed for Ki67 as part of the antigen retrieval process using the low pH EnVision FLEX Target Retrieval Solutions (Dako) for 20 min at 97C using a PT Link (Dako, Agilent). For caspase 3, samples were dewaxed, and antigen retrieval was performed with citrate buffer (pH 6) for 20 min at 121C with an autoclave. Endogenous peroxidase was quenched by 10-min incubation with peroxidase blocking solution (Dako REAL, S2023). The rabbit polyclonal primary antibodies anti-Ki67 (Abcam, ab15580) and anti-cleaved caspase 3 (Cell Signaling Technology, 9661S) were diluted 1:1000 and 1:300, respectively, with EnVision FLEX Antibody Diluent (Dako, Agilent, K800621) and incubated for 60 and 120 min, respectively, at room temperature. A biotin-free, ready-to-use BrightVision polyhorseradish peroxidase (HRP)anti-rabbit immunoglobulin G (Immunologic, DPVR-110HRP) was used as secondary antibody. Immunohistochemistry for ER (clone 1D5; Dako, M7047), PR (Abcam, ab63605), and RANKL (R&D Systems, AF462) was performed as previously described (61, 62). Antigen-antibody complexes were revealed with 3,30-diaminobenzidine tetrahydrochloride (Dako, K3468). Sections were counterstained with hematoxylin (Dako, S202084) and mounted with toluene-free mounting medium (Dako, CS705) using a Dako CoverStainer. Bright-field images were acquired with a NanoZoomer-2.0 HT C9600 scanner (Hamamatsu). All images were visualized with a gamma correction set at 1.8 in the image control panel of the NDP.view software (Hamamatsu, Photonics, France). Image analysis was performed using TMARKER software (63). For immunofluorescence, Alexa secondary antibodies and 4,6-diamidino-2-phenylindole (DAPI) were used, and images were obtained on an inverted Leica TCS SP5 confocal microscopy.

Beads-homogenized tissue or MECs (EasySep, STEMCELL Technologies) were lysed in ice-cold radioimmunoprecipitation assay (RIPA) lysis buffer (with phosphatase and protease inhibitors) and sonicated for 5 min at high or low intensity, respectively (Standard Bioruptor Diagenode). Cellular debris was pelleted (15,700g, 15 min, 4C), and protein concentration was determined by the DC Protein Assay (Bio-Rad). Equal amounts of proteins were separated by SDSpolyacrylamide gel electrophoresis. After transfer onto nitrocellulose membranes (Sigma-Aldrich, GE10600001), membranes were blocked for 1 hour in 5% milk, and specific proteins were labeled with the corresponding primary antibodies against vinculin (Abcam, ab18058), CPEB3 (Abcam, ab10883), CPEB219, CPEB4 (Abcam, ab83009), CPEB1 (Cell Signaling Technology, no. 13583), CyclinD1 (Santa Cruz Biotechnology, sc-717), CREB1 (Cell Signaling Technology, no. 9197), -tubulin (Sigma-Aldrich, T9026), and glyceraldehyde-3-phosphate dehydrogenase (GAPDH; Life Technologies, AM-4300). Secondary HRP antibodies were also diluted in 5% milk, and proteins were revealed using enhanced chemiluminescence Western blotting detection reagents (GE Healthcare).

Human breast carcinoma cell lines MDA-MB-231, BT549, MDA-MB-435, MDA-MB-468, SKBR3, BT474, T47D, MCF7, and ZR75 were obtained from the American Type Culture CollectionLGC Standards Ltd. Partnership. All cell lines were cultured in DMEM d-glucose medium (Gibco) supplemented with 10% fetal bovine serum and 1% penicillin/streptomycin, except BT459 cells, which were cultured in supplemented RPMI medium (Gibco). All cells were cultured at 37C and in a 5% CO2 humidified atmosphere. For lentiviral infection, human embryonic kidney293 T cells were transfected with pLKO lentiviral vectors and plasmids encoding lentiviral particles using standard methods. pLKO sh_CPEB2 plasmids were obtained from Sigma-Aldrich MISSION shRNA library (clones TRCN0000149728 and TRCN0000149778). Recipient cells were transduced with the viral medium and selected with puromycin (2 g ml1) for 72 hours.

In vitro cell proliferation was assessed using the CyQUANT Cell Proliferation Kit following the manufacturers instructions. For 4-OHT sensitivity experiments, 4-OHT or vehicle (ethanol) was added to the cell culture at the indicated concentrations 24 hours after plating. Cell numbers were quantified after 6 days using BIO-TEK FL600 fluorescence microplate reader at 485 to 530 nm.

To detect early apoptosis (APC labeled), cultured cells were trypsinized and processed following the Annexin V Apoptosis Detection Kit (Thermo Fisher Scientific). DAPI solution was also added to the cell suspension to detect the total number of dead cells. A Gallios cytometer (Beckman Coulter) was used for the analysis.

Total RNA was extracted by TRIzol reagent (Invitrogen). RNA (1 g) was reverse-transcribed with oligo(dT) and random primers using SuperScript IV (Thermo Fisher Scientific) or RevertAid (Thermo Fisher Scientific), following the manufacturers recommendations. Real-time qPCR (RT-qPCR) was performed in a LightCycler 480 (Roche) using PowerUp SYBR Green Master Mix (Roche). Primer sequences are listed in table S2. RNA quantifications were normalized to GAPDH as endogenous control. For human breast carcinoma cell lines, RNA extraction (PureLink RNA Mini Kit, Thermo Fisher Scientific), reverse transcription (High-Capacity cDNA Reverse Transcription Kit, Applied Biosystems), and real-time PCR (TaqMan Universal Master Mix, Applied Biosystems) were performed and analyzed as previously described (64). The TaqMan probes (Applied Biosystems) used were Hs0139673_m1 (CPEB2), Hs00153408_m1 (MYC), Hs00765553_m1 (CCND1), and Mm00437762_m1 (B2M). For microarrays, samples in duplicates from sorted cells from WT and CPEB2 KO animals were processed at IRB Barcelonas Functional Genomics Core Facility following standard procedures. Affymetrix MG-430 PM strip data for DPs, DD, APs, and myoepithelial cell population samples in WT and CPEB2 KO in biological duplicates were processed with Bioconductor (65) using robust multiarray average (RMA) background correction, quantile normalization, and RMA summarization to obtain probeset expression estimates (66). Centroid locations from the principal component for the different combinations between cell populations and genotypes, as well as the resultant Euclidean distances between centroids, were computed. Dispersion within groups (the average Euclidean distance between samples and their corresponding population/genotype centroid) was also measured. Limma 3.22.7 (67) was then used to identify differentially expressed genes between CPEB2 KO and WT in all four cell populations, with P < 0.01 and |FC| > 2. Lists of up- and down-regulated genes between DPWT and DDWT were generated by selecting candidate genes with the highest and lowest FC percentiles and P < 0.01 (1% most up- and down-regulated genes, n = 181 and n = 101, respectively). Alternatively, after selecting with the highest and lowest FC percentiles, we also filtered these using a FDR threshold of 0.1. This resulted in a more stringent list of 24 up-regulated and no down-regulated genes in WT DP versus WT NCL. Enrichment for these gene lists, as well as for Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Broad Institute hallmark gene set categories in whole-genome gene lists ranked by mean log2FC between cell populations and genotypes, was assessed with the GSEA preranked algorithm (68). M. musculus GO and KEGG gene set collections were generated using the org.Mm.eg.db Bioconductor package (October 2014). Homo sapiens Hallmark gene set was downloaded from the Molecular Signatures Database and translated to M. musculus using Ensembl human-mouse homology information (August 2016).

MECs (EasySep, STEMCELL Technologies) were isolated from WT and CPEB2 KO animals (with two animals pooled per duplicate). Pellets were washed twice with cold Hanks balanced salt solution, lysed with RIPA buffer [50 mM tris-HCl (pH 8), 150 mM NaCl, 1 mM MgCl2, 1% NP-40, 1 mM EDTA, 0.1% SDS, protease inhibitor cocktail, and ribonuclease inhibitors] and sonicated for 5 min at low intensity with Standard Bioruptor Diagenode. After centrifugation (10 min, 4C), supernatants were collected, precleared, and immunoprecipitated (4 hours, 4C) with 10 g of anti-CPEB2 antibody (69) bound to 50 l of Dynabeads Protein G (Invitrogen). Beads were washed and split for either protein or RNA extraction. For RNA isolation, beads were resuspended in 100 l of proteinase K buffer with 70 g of proteinase K (Roche) and incubated for 30 min at 42C and 30 min at 65C. RNA was extracted following standard phenol/chloroform protocol. Samples were processed at IRB Barcelonas Functional Genomics Facility following standard procedures: Illumina Hi-Seq 2000 50base pair single-end RIP-sequencing (RIP-seq) data for WT and CPEB2 KO in biological duplicates, as well as their respective input samples of MECs, were checked for general sequencing quality control and adapter contamination using the FastQC software version 0.11, and no relevant problems were found. Afterward, reads were aligned against the M. musculus University of California, Santa Cruz mm10 ribosomal RNA (rRNA) genome using Bowtie1 0.12.9 (70) with two mismatches and default options to identify and remove reads coming from potential rRNA contamination from downstream analysis. Curated (non-rRNA) reads were then aligned against the M. musculus mm10 reference genome using Bowtie2 2.2.2 (71), allowing for one mismatch and reporting the best alignment site per read. All samples reported >15 million aligned reads. Potential amplification artefacts (duplicated reads) were detected and removed with the sambamba software version 0.5.1 using default options. Binary tiled data file tracks for visual inspection in the Integrative Genomics Viewer (IGV) software were generated using igvtools version 2. Read counts at 3UTR level (longest 3UTR per gene, mm10 genome Ensembl, March 2017) were computed using the featureCounts function from the Rsubread package version 1.24.2 with options minMQS = 1. Then, an interaction analysis of WT and CPEB2 KO RIP samples and their respective input controls (RIPWT/InputWT versus RIPKO/InputKO) was performed with DESeq2 (72). Target 3UTRs were selected using an interaction FC threshold of >1.5 and interaction Benjamini-Hochberg adjusted P < 0.1 (see table S1, high-confidence RIP target genes, n = 169). GO enrichment for selected targets was performed using the online Enrichr (73, 74) tool.

For animal experiments, data were expressed as means SEM, and statistics were analyzed with the GraphPad Prism software. Experiments were performed following a randomized block design. Littermates kept in the same cage since weaning were used whenever possible. The experiment was blinded before experimental analysis. For human breast carcinoma cell lines, P values were generated using the Students t test (unpaired, two tailed); P < 0.05 was considered significant. Error bars were calculated as SE in all the statistical analysis shown. Number of independent experiments is indicated in the figure legends.

Transcriptomic and clinical data from the METABRIC breast cancer dataset (75, 76) were downloaded from the cBioPortal for Cancer Genomics database (77). Association of gene expression with molecular features (PAM50 subtype and ER status) was evaluated using a linear model, while a Cox model was fitted to assess association with overall survival. Statistical significance was assessed using the corresponding F tests of log-likelihood ratio tests. A Wald test was used for pairwise comparisons when necessary. In all cases, the cohort of origin of the sample was included as a covariate in the models.

For survival analyses, sample groups of low, medium, and high expression levels were defined using the tertiles of the intensity distribution after correction by cohort effects, as estimated by a linear model in which PAM50 subtypes were included as covariates. Association of gene expression with early relapse was modeled using a step function for a prespecified cutoff of a 10-year follow-up. Hazard ratios and their corresponding 95% confidence intervals were computed as a measure of association. For visualization purposes, Kaplan-Meier curves were estimated for groups of tumors that showed low, medium, or high expression. The threshold for statistical significance was set at 5%. All analyses were conducted with R (78).

Follow this link:
The RNA binding protein CPEB2 regulates hormone sensing in mammary gland development and luminal breast cancer - Science Advances

Stem Cells Market Market Developments and Analysis (impact of Covid-19) 2020-2026 – Herald Writeup

Stem Cells Market research report provides an actual industry viewpoint, future trends and dynamics for market growth rate, market size, trading and key players of the industry with forecast period of 2026. This comprehensive research report is titled Stem Cells Market with Industry Analysis and Opportunity Assessment and it comprises a whole market scenario along with the dynamics affecting it.

Inquire for Sample Copy of this Report : https://www.marketinsightsreports.com/reports/05152017541/global-stem-cells-market-report-2020-by-key-players-types-applications-countries-market-size-forecast-to-2026-based-on-2020-covid-19-worldwide-spread/inquiry?heraldwriteup

The report presents the market competitive landscape and a corresponding detailed analysis of the major vendor/key players in the market. Top Companies in the Global Stem Cells Market: Apceth GmbH?Company KGNeostem Oncology, LlcMesoblastBiotime, Inc.Ocata Therapeutics, Inc.Pharmicell Co., Ltd.Gamida Cell Ltd.U.S. Stem Cell, Inc. (Bioheart)Cell Cure Neurosciences Ltd.Stemcells, Inc.Medipost Co., Ltd.Pluristem Therapeutics Inc.Reneuron Group PlcNeuralstem, Inc.Stempeutics Research Pvt. Ltd.Orthocyte CorporationAnterogen Co., Ltd.

and others.

Global Stem Cells Market Split by Product Type and Applications:

This report segments the global Stem Cells market on the basis ofTypesare:Adult Stem CellHuman Embryonic Stem CellInduced Pluripotent Stem CellRat Neural Stem CellOther

On the basis of Application, the Global Stem Cells market is segmented into:Drug Discovery and DevelopmentStem Cells Regenerative Medicine

Regional Analysis For Stem Cells Market:

North America (United States, Canada and Mexico)Europe (Germany, France, UK, Russia and Italy)Asia-Pacific (China, Japan, Korea, India and Southeast Asia)South America (Brazil, Argentina, Colombia etc.)Middle East and Africa (Saudi Arabia, UAE, Egypt, Nigeria and South Africa)

Browse the report description and TOC:

https://www.marketinsightsreports.com/reports/05152017541/global-stem-cells-market-report-2020-by-key-players-types-applications-countries-market-size-forecast-to-2026-based-on-2020-covid-19-worldwide-spread?heraldwriteup

Influence of the Stem Cells Market Report:

-Comprehensive assessment of all opportunities and risk in the Stem Cells market.-Detailed study of business strategies for growth of the Stem Cells market-leading players.-Conclusive study about the growth plot of Stem Cells market for forthcoming years.-In-depth understanding of Stem Cells market-particular drivers, constraints and major micro markets.-Favourable impression inside vital technological and market latest trends striking the Stem Cells market.

Get Flat 20% Discount at: https://www.marketinsightsreports.com/reports/05152017541/global-stem-cells-market-report-2020-by-key-players-types-applications-countries-market-size-forecast-to-2026-based-on-2020-covid-19-worldwide-spread/discount?heraldwriteup

Key Strategic Developments: The study also includes the key strategic developments of the market, comprising, new product launch, agreements, collaborations, partnerships, joint ventures, and regional growth of the leading competitors operating in the market on a global and regional scale.

Key Market Features: The report evaluated key market features, including revenue, price, capacity, capacity utilisation rate, gross, production, production rate, consumption, import/export, supply/demand, cost, market share, CAGR, and gross margin. In addition, the study offers a comprehensive study of the key market dynamics and their latest trends, along with pertinent market segments and sub-segments.

Analytical Tools: The Global Stem Cells Market report includes the accurately studied and assessed data of the key industry players and their scope in the market by means of a number of analytical tools. The analytical tools such as Porters five forces analysis, SWOT analysis, feasibility study, and investment return analysis have been used to analyse the growth of the key players operating in the market.

Customisation of the Report: This report can be customised as per your needs for additional data up to 3 companies or countries or 40 analyst hours.

Please connect with our sales team ([emailprotected] ).

About Us:

MarketInsightsReports provides syndicated market research on industry verticals including Healthcare, Information and Communication Technology (ICT), Technology and Media, Chemicals, Materials, Energy, Heavy Industry, etc.MarketInsightsReports provides global and regional market intelligence coverage, a 360-degree market view which includes statistical forecasts, competitive landscape, detailed segmentation, key trends, and strategic recommendations.

Contact Us:

Irfan Tamboli (Head of Sales) Market Insights Reports

Phone: + 1704 266 3234 | +91-750-707-8687

[emailprotected] | [emailprotected]

This document has been composed with the free online HTML converter. Click here to give it a try.

https://www.linkedin.com/company/13411016/https://twitter.com/MIRresearch

See the rest here:
Stem Cells Market Market Developments and Analysis (impact of Covid-19) 2020-2026 - Herald Writeup

Takeda Pharmaceutical : China announces ADCETRIS (brentuximab vedotin) is approved for the treatment of adult patients with CD30-positive Lymphomas -…

- Approval will provide new treatment option in China for patients diagnosed with relapsed or refractory system anaplastic large cell lymphoma (sALCL) or Hodgkin lymphoma- Takeda China committed to the continued delivery of highly innovative medicines to patients

Shanghai, CHINA and Osaka, JAPAN May 15, 2020 - Takeda China announced today that ADCETRIS (brentuximab vedotin) has been officially approved by China's National Medical Products Administration (NMPA) for use in adult patients with relapsed or refractory systemic Anaplastic Large Cell Lymphoma (sALCL) or CD30-positive Hodgkin Lymphoma.

'We expect that brentuximab vedotin will provide a better treatment option for CD30-positive lymphoma in China,' said Professor Zhu Jun, Director of the Lymphoma Department at Beijing Cancer Hospital and Principal Investigator of the ADCETRIS registration study in China. 'Both sALCL and classical Hodgkin lymphoma are subtypes of lymphoma that express CD30. For decades, treatment options for patients in China with relapsed or refractory lymphoma have been very limited. Patients' overall survival rates are low, and their quality of life is also negatively affected.'

Lymphoma is a type of malignant tumor that originates in the lymphohematopoietic system. It's the collective name of more than 70 subtypes in the lymphoma family[1]. It is one of the ten most malignant cancers in China with the highest mortality rates. Data shows that each year in China, approximately 93,000 people are diagnosed with lymphoma, and more than 50,000 people die from it[2]. Currently there are very limited therapies available for treating patients with relapsed or refractory lymphoma in China.

'ADCETRIS was granted priority review by the Center for Drug Evaluation in June 2019 and has now been officially approved by the NMPA. This 'fast-track' approval process demonstrates the Chinese government's determination to accelerate the introduction of highly innovative drugs to China's patients. We are thankful for their accelerated approval and the hope it gives to patients with relapsed or refractory lymphoma,' said Sean Shan, President of Takeda China. 'As Takeda aims to put the patient at the center of everything we do, we are committed to leveraging our global R&D capabilities and local operations to accelerate the pace at which we bring innovative drugs to address the unmet needs of patients in China and support the government's 'Healthy China 2030' initiative.'

[1] WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues (2016)

[2] The Global Cancer Observatory. China factsheets. 2019.

About ADCETRISADCETRIS is an antibody-drug conjugate (ADC) comprising an anti-CD30 monoclonal antibody attached by a protease-cleavable linker to a microtubule disrupting agent, monomethyl auristatin E (MMAE), utilizing Seattle Genetics' proprietary technology. The ADC employs a linker system that is designed to be stable in the bloodstream but to release MMAE upon internalization into CD30-positive tumor cells.

ADCETRIS injection for intravenous infusion has received FDA approval for six indications in adult patients with: (1) previously untreated systemic anaplastic large cell lymphoma (sALCL) or other CD30-expressing peripheral T-cell lymphomas (PTCL), including angioimmunoblastic T-cell lymphoma and PTCL not otherwise specified, in combination with cyclophosphamide, doxorubicin, and prednisone, (2) previously untreated Stage III or IV classical Hodgkin lymphoma (cHL), in combination with doxorubicin, vinblastine, and dacarbazine, (3) cHL at high risk of relapse or progression as post-autologous hematopoietic stem cell transplantation (auto-HSCT) consolidation, (4) cHL after failure of auto-HSCT or failure of at least two prior multi-agent chemotherapy regimens in patients who are not auto-HSCT candidates, (5) sALCL after failure of at least one prior multi-agent chemotherapy regimen, and (6) primary cutaneous anaplastic large cell lymphoma (pcALCL) or CD30-expressing mycosis fungoides (MF) who have received prior systemic therapy.

Health Canada granted ADCETRIS approval with conditions for relapsed or refractory Hodgkin lymphoma and sALCL in 2013, and non-conditional approval for post-autologous stem cell transplantation (ASCT) consolidation treatment of Hodgkin lymphoma patients at increased risk of relapse or progression in 2017, adults with pcALCL or CD30-expressing MF who have had prior systemic therapy in 2018, for previously untreated Stage IV Hodgkin lymphoma in combination with doxorubicin, vinblastine, and dacarbazine in 2019 and for previously untreated adult patients with sALCL, peripheral T-cell lymphoma-not otherwise specified (PTCL-NOS) or angioimmunoblastic T-cell lymphoma (AITL), whose tumors express CD30, in combination with cyclophosphamide, doxorubicin, prednisone in 2019.

ADCETRIS received conditional marketing authorization from the European Commission in October 2012. The approved indications in Europe are: (1) for the treatment of adult patients with previously untreated CD30-positive Stage IV Hodgkin lymphoma in combination with doxorubicin, vinblastine and dacarbazine (AVD), (2) for the treatment of adult patients with CD30-positive Hodgkin lymphoma at increased risk of relapse or progression following ASCT, (3) for the treatment of adult patients with relapsed or refractory CD30-positive Hodgkin lymphoma following ASCT, or following at least two prior therapies when ASCT or multi-agent chemotherapy is not a treatment option, (4) for the treatment of adult patients with relapsed or refractory sALCL and (5) for the treatment of adult patients with CD30-positive cutaneous T-cell lymphoma (CTCL) after at least one prior systemic therapy.

In Japan, ADCETRIS received its first approval in January 2014 for relapsed or refractory Hodgkin lymphoma and ALCL, and untreated Hodgkin lymphoma in combination with doxorubicin, vinblastine, and dacarbazine in September 2018, and Peripheral T-cell lymphomas in December 2019. In December 2019, ADCETRIS obtained additional dosage & administration for the treatment of relapsed or refractory Hodgkin lymphoma and Peripheral T-cell lymphomas in pediatric. The current wording of approved indication in Japan package insert is for the treatment of patients with CD30 positive: Hodgkin lymphoma and Peripheral T-cell lymphomas.

ADCETRIS has received marketing authorization by regulatory authorities in more than 70 countries/ regions for relapsed or refractory Hodgkin lymphoma and sALCL. See important safety information below.

ADCETRIS is being evaluated broadly in more than 70 clinical trials, including a Phase 3 study in first-line Hodgkin lymphoma (ECHELON-1) and another Phase 3 study in first-line CD30-positive peripheral T-cell lymphomas (ECHELON-2), as well as trials in many additional types of CD30-positive malignancies.

Seattle Genetics and Takeda are jointly developing ADCETRIS. Under the terms of the collaboration agreement, Seattle Genetics has U.S. and Canadian commercialization rights and Takeda has rights to commercialize ADCETRIS in the rest of the world. Seattle Genetics and Takeda are funding joint development costs for ADCETRIS on a 50:50 basis, except in Japan where Takeda is solely responsible for development costs.

ADCETRIS Approval in ChinaThe local approval of ADCETRIS in China is based on the data from clinical studies SG035-0004, SG035-0003 and C25007. In the SG-035-0004 study, among 58 patients with relapsed or refractory sALCL, tumor reduction was seen in 97%[3], and their five-year survival rate was 60%[4]. In the SG-035-0003 study, among 102 patients with relapsed or refractory cHL, tumor reduction was seen in 94%[5], with the median overall survival (OS) increasing from a historical 27.6 months to 40.5 months[6]. The C25007 study is a Phase IV single-arm study of patients (n=60) with relapsed or refractory cHL who had received chemotherapy at least once and were not suitable for stem cell transplantation (SCT), or multidrug chemotherapy when they began to receive treatment with brentuximab vedotin. In this study, the objective remission rate for the subjects was 50% (95% CI, 37: 63%)[7].

[3] Pro, B., et al. Brentuximab Vedotin (SGN-35) in Patients With Relapsed or Refractory Systemic Anaplastic Large-Cell Lymphoma: Results of a Phase II Study. Journal of Clinical Oncology 2012 30:18, 2190-2196.

[4] Pro, B., et al. Five-year results of brentuximab vedotin in patients with relapsed or refractory systemic anaplastic large cell lymphoma. Blood vol. 130,25 (2017): 2709-2717.

[5] Younes, A., et al. Results of a Pivotal Phase II Study of Brentuximab Vedotin for Patients With Relapsed or Refractory Hodgkin's Lymphoma. Journal of Clinical Oncology 2012 30:18, 2183-2189.

[6] Chen R., et al. Five-year survival and durability results of brentuximab vedotin in patients with relapsed or refractory Hodgkin lymphoma. Blood 2016; 128 (12): 1562-1566.

[7] Canadian Agency for Drugs and Technologies in Health. Brentuximab (Adcetris) for Hodgkin Lymphoma - Resubmission. 2019.

ADCETRIS (brentuximab vedotin) Important Safety Information (European Union)Please refer to Summary of Product Characteristics (SmPC) before prescribing.

CONTRAINDICATIONS

ADCETRIS is contraindicated for patients with hypersensitivity to brentuximab vedotin and its excipients. In addition, combined use of ADCETRIS with bleomycin causes pulmonary toxicity.

SPECIAL WARNINGS & PRECAUTIONS

Progressive multifocal leukoencephalopathy (PML): John Cunningham virus (JCV) reactivation resulting in progressive multifocal leukoencephalopathy (PML) and death can occur in patients treated with ADCETRIS. PML has been reported in patients who received ADCETRIS after receiving multiple prior chemotherapy regimens. PML is a rare demyelinating disease of the central nervous system that results from reactivation of latent JCV and is often fatal.

Closely monitor patients for new or worsening neurological, cognitive, or behavioral signs or symptoms, which may be suggestive of PML. Suggested evaluation of PML includes neurology consultation, gadolinium-enhanced magnetic resonance imaging of the brain, and cerebrospinal fluid analysis for JCV DNA by polymerase chain reaction or a brain biopsy with evidence of JCV. A negative JCV PCR does not exclude PML. Additional follow up and evaluation may be warranted if no alternative diagnosis can be established. Hold dosing for any suspected case of PML and permanently discontinue ADCETRIS if a diagnosis of PML is confirmed.

Be alert to PML symptoms that the patient may not notice (e.g., cognitive, neurological, or psychiatric symptoms).

Pancreatitis: Acute pancreatitis has been observed in patients treated with ADCETRIS. Fatal outcomes have been reported. Closely monitor patients for new or worsening abdominal pain, which may be suggestive of acute pancreatitis. Patient evaluation may include physical examination, laboratory evaluation for serum amylase and serum lipase, and abdominal imaging, such as ultrasound and other appropriate diagnostic measures. Hold ADCETRIS for any suspected case of acute pancreatitis. ADCETRIS should be discontinued if a diagnosis of acute pancreatitis is confirmed.

Pulmonary Toxicity: Cases of pulmonary toxicity, some with fatal outcomes, including pneumonitis, interstitial lung disease, and acute respiratory distress syndrome (ARDS), have been reported in patients receiving ADCETRIS. Although a causal association with ADCETRIS has not been established, the risk of pulmonary toxicity cannot be ruled out. Promptly evaluate and treat new or worsening pulmonary symptoms (e.g., cough, dyspnoea) appropriately. Consider holding dosing during evaluation and until symptomatic improvement.

Serious infections and opportunistic infections: Serious infections such as pneumonia, staphylococcal bacteremia, sepsis/septic shock (including fatal outcomes), and herpes zoster, cytomegalovirus (CMV) (reactivation) and opportunistic infections such as Pneumocystis jiroveci pneumonia and oral candidiasis have been reported in patients treated with ADCETRIS. Patients should be carefully monitored patients during treatment for the emergence of possible serious and opportunistic infections.

Infusion-related reactions (IRR): Immediate and delayed IRR, as well as anaphylaxis, have been reported with ADCETRIS. Carefully monitor patients during and after an infusion. If anaphylaxis occurs, immediately and permanently discontinue administration of ADCETRIS and administer appropriate medical therapy. If an IRR occurs, interrupt the infusion and institute appropriate medical management. The infusion may be restarted at a slower rate after symptom resolution. Patients who have experienced a prior IRR should be premedicated for subsequent infusions. IRRs are more frequent and more severe in patients with antibodies to ADCETRIS.

Tumor lysis syndrome (TLS): TLS has been reported with ADCETRIS. Patients with rapidly proliferating tumor and high tumor burden are at risk of TLS. Monitor these patients closely and manage according to best medical practice.

Peripheral neuropathy (PN): ADCETRIS treatment may cause PN, both sensory and motor. ADCETRIS-induced PN is typically an effect of cumulative exposure to ADCETRIS and is reversible in most cases. Monitor patients for symptoms of neuropathy, such as hypoesthesia, hyperesthesia, paresthesia, discomfort, a burning sensation, neuropathic pain, or weakness. Patients experiencing new or worsening PN may require a delay and a dose reduction or discontinuation of ADCETRIS.

Hematological toxicities: Grade 3 or Grade 4 anemia, thrombocytopenia, and prolonged (equal to or greater than one week) Grade 3 or Grade 4 neutropenia can occur with ADCETRIS. Monitor complete blood counts prior to administration of each dose.

Febrile neutropenia: Febrile neutropenia has been reported with ADCETRIS. Complete blood counts should be monitored prior to administration of each dose of treatment. Closely monitor patients for fever and manage according to best medical practice if febrile neutropenia develops.When ADCETRIS is administered in combination with AVD, primary prophylaxis with G-CSF is recommended for all patients beginning with the first dose.

Stevens-Johnson syndrome (SJS): SJS and toxic epidermal necrolysis (TEN) have been reported with ADCETRIS. Fatal outcomes have been reported. Discontinue treatment with ADCETRIS if SJS or TEN occurs and administer appropriate medical therapy.

Gastrointestinal (GI) Complications: GI complications, some with fatal outcomes, including intestinal obstruction, ileus, enterocolitis, neutropenic colitis, erosion, ulcer, perforation and haemorrhage, have been reported with ADCETRIS. Promptly evaluate and treat patients if new or worsening GI symptoms occur.

Hepatotoxicity: Elevations in alanine aminotransferase (ALT) and aspartate aminotransferase (AST) have been reported with ADCETRIS. Serious cases of hepatotoxicity, including fatal outcomes, have also occurred. Pre-existing liver disease, comorbidities, and concomitant medications may also increase the risk. Test liver function prior to treatment initiation and routinely monitor during treatment. Patients experiencing hepatotoxicity may require a delay, dose modification, or discontinuation of ADCETRIS.

Hyperglycemia: Hyperglycemia has been reported during trials in patients with an elevated body mass index (BMI) with or without a history of diabetes mellitus. Closely monitor serum glucose for patients who experiences an event of hyperglycemia. Administer anti-diabetic treatment as appropriate.

Renal and Hepatic Impairment: There is limited experience in patients with renal and hepatic impairment. Available data indicate that MMAE clearance might be affected by severe renal impairment, hepatic impairment, and by low serum albumin concentrations.

CD30+ CTCL: The size of the treatment effect in CD30 + CTCL subtypes other than mycosis fungoides (MF) and primary cutaneous anaplastic large cell lymphoma (pcALCL) is not clear due to lack of high level evidence. In two single arm phase II studies of ADCETRIS, disease activity has been shown in the subtypes Szary syndrome (SS), lymphomatoid papulosis (LyP) and mixed CTCL histology. These data suggest that efficacy and safety can be extrapolated to other CTCL CD30+ subtypes. Carefully consider the benefit-risk per patient and use with caution in other CD30+ CTCL patient types.

Sodium content in excipients: This medicinal product contains 13.2 mg sodium per vial, equivalent to 0.7% of the WHO recommended maximum daily intake of 2 g sodium for an adult.

INTERACTIONSPatients who are receiving a strong CYP3A4 and P-gp inhibitor, concomitantly with ADCETRIS may have an increased risk of neutropenia. If neutropenia develops, refer to dosing recommendations for neutropenia (see SmPC section 4.2). Co-administration of ADCETRIS with a CYP3A4 inducer did not alter the plasma exposure of ADCETRIS, but it appeared to reduce plasma concentrations of MMAE metabolites that could be assayed. ADCETRIS is not expected to alter the exposure to drugs that are metabolized by CYP3A4 enzymes.

PREGNANCY: Advise women of childbearing potential to use two methods of effective contraception during treatment with ADCETRIS and until 6 months after treatment. There are no data from the use of ADCETRIS in pregnant women, although studies in animals have shown reproductive toxicity. Do not use ADCETRIS during pregnancy unless the benefit to the mother outweighs the potential risks to the fetus.

LACTATION (breast-feeding): There are no data as to whether ADCETRIS or its metabolites are excreted in human milk, therefore a risk to the newborn/infant cannot be excluded. With the potential risk, a decision should be made whether to discontinue breast-feeding or discontinue/abstain from therapy with ADCETRIS.

FERTILITY: In nonclinical studies, ADCETRIS treatment has resulted in testicular toxicity, and may alter male fertility. Advise men being treated with ADCETRIS not to father a child during treatment and for up to 6 months following the last dose.

Effects on ability to drive and use machines: ADCETRIS may have a moderate influence on the ability to drive and use machines.

UNDESIRABLE EFFECTS

Monotherapy: The most frequent adverse reactions (10%) were infections, peripheral sensory neuropathy, nausea, fatigue, diarrhoea, pyrexia, upper respiratory tract infection, neutropenia, rash, cough, vomiting, arthralgia, peripheral motor neuropathy, infusion-related reactions, pruritus, constipation, dyspnoea, weight decreased, myalgia and abdominal pain. Serious adverse drug reactions occurred in 12% of patients. The frequency of unique serious adverse drug reactions was 1%. Adverse events led to treatment discontinuation in 24% of patients.

Combination Therapy: In the study of ADCETRIS as combination therapy with AVD in 662 patients with previously untreated advanced HL, the most common adverse reactions ( 10%) were: neutropenia, nausea, constipation, vomiting, fatigue, peripheral sensory neuropathy, diarrhoea, pyrexia, alopecia, peripheral motor neuropathy, decreased weight, abdominal pain, anaemia, stomatitis, febrile neutropenia, bone pain, insomnia, decreased appetite, cough, headache, arthralgia, back pain, dyspnoea, myalgia, upper respiratory tract infection, alanine aminotransferase increased. Serious adverse reactions occurred in 36% of patients. Serious adverse reactions occurring in 3% of patients included febrile neutropenia (17%), pyrexia (6%), and neutropenia (3%). Adverse events led to treatment discontinuation in 13% of patients.

ADCETRIS (brentuximab vedotin) U.S. Important Safety Information

BOXED WARNINGPROGRESSIVE MULTIFOCAL LEUKOENCEPHALOPATHY (PML): JC virus infectionresulting in PML and death can occur in ADCETRIS-treated patients.

ContraindicationADCETRIS concomitant with bleomycin due to pulmonary toxicity (e.g., interstitial infiltration and/or inflammation).

Warnings and Precautions

Administer G-CSF primary prophylaxis beginning with Cycle 1 for patients who receive ADCETRIS in combination with chemotherapy for previously untreated Stage III/IV cHL or previously untreated PTCL.

Monitor complete blood counts prior to each ADCETRIS dose. Monitor more frequently for patients with Grade 3 or 4 neutropenia. Monitor patients for fever. If Grade 3 or 4 neutropenia develops, consider dose delays, reductions, discontinuation, or G-CSF prophylaxis with subsequent doses.

Most Common (20% in any study) Adverse ReactionsPeripheral neuropathy, fatigue, nausea, diarrhea, neutropenia, upper respiratory tract infection, pyrexia, constipation, vomiting, alopecia, decreased weight, abdominal pain, anemia, stomatitis, lymphopenia, and mucositis.

Drug InteractionsConcomitant use of strong CYP3A4 inhibitors or inducers has the potential to affect the exposure to monomethyl auristatin E (MMAE).

Use in Specific PopulationsModerate or severe hepatic impairment or severe renal impairment: MMAE exposure and adverse reactions are increased. Avoid use.

Advise males with female sexual partners of reproductive potential to use effective contraception during ADCETRIS treatment and for at least 6 months after the final dose of ADCETRIS.

Advise patients to report pregnancy immediately and avoid breastfeeding while receiving ADCETRIS.

Please see the full Prescribing Information, including BOXED WARNING, for ADCETRIS here

About Takeda Pharmaceutical Company LimitedTakeda Pharmaceutical Company Limited (TSE:4502/NYSE:TAK) is a global, values-based, R&D-driven biopharmaceutical leader headquartered in Japan, committed to bringing Better Health and a Brighter Future to patients by translating science into highly-innovative medicines. Takeda focuses its R&D efforts on four therapeutic areas: Oncology, Rare Diseases, Neuroscience, and Gastroenterology (GI). We also make targeted R&D investments in Plasma-Derived Therapies and Vaccines. We are focusing on developing highly innovative medicines that contribute to making a difference in people's lives by advancing the frontier of new treatment options and leveraging our enhanced collaborative R&D engine and capabilities to create a robust, modality-diverse pipeline. Our employees are committed to improving quality of life for patients and to working with our partners in health care in approximately 80 countries.For more information, visit https://www.takeda.com.

Media Contacts:

Japanese MediaKazumi Kobayashikazumi.kobayashi@takeda.com+81 (0) 3-3278-2095

Media outside JapanSara Noonansara.noonan@takeda.com+1-617-551-3683

Lynette Chenglynette.cheng@takeda.com+65-9853-6628

Important NoticeFor the purposes of this notice, 'press release' means this document, any oral presentation, any question and answer session and any written or oral material discussed or distributed by Takeda Pharmaceutical Company Limited ('Takeda') regarding this release. This press release (including any oral briefing and any question-and-answer in connection with it) is not intended to, and does not constitute, represent or form part of any offer, invitation or solicitation of any offer to purchase, otherwise acquire, subscribe for, exchange, sell or otherwise dispose of, any securities or the solicitation of any vote or approval in any jurisdiction. No shares or other securities are being offered to the public by means of this press release. No offering of securities shall be made in the United States except pursuant to registration under the U.S. Securities Act of 1933, as amended, or an exemption therefrom. This press release is being given (together with any further information which may be provided to the recipient) on the condition that it is for use by the recipient for information purposes only (and not for the evaluation of any investment, acquisition, disposal or any other transaction). Any failure to comply with these restrictions may constitute a violation of applicable securities laws.The companies in which Takeda directly and indirectly owns investments are separate entities. In this press release, 'Takeda' is sometimes used for convenience where references are made to Takeda and its subsidiaries in general. Likewise, the words 'we', 'us' and 'our' are also used to refer to subsidiaries in general or to those who work for them. These expressions are also used where no useful purpose is served by identifying the particular company or companies.

Forward-Looking StatementsThis press release and any materials distributed in connection with this press release may contain forward-looking statements, beliefs or opinions regarding Takeda's future business, future position and results of operations, including estimates, forecasts, targets and plans for Takeda. Without limitation, forward-looking statements often include words such as 'targets', 'plans', 'believes', 'hopes', 'continues', 'expects', 'aims', 'intends', 'ensures', 'will', 'may', 'should', 'would', 'could' 'anticipates', 'estimates', 'projects' or similar expressions or the negative thereof. These forward-looking statements are based on assumptions about many important factors, including the following, which could cause actual results to differ materially from those expressed or implied by the forward-looking statements: the economic circumstances surrounding Takeda's global business, including general economic conditions in Japan and the United States; competitive pressures and developments; changes to applicable laws and regulations; the success of or failure of product development programs; decisions of regulatory authorities and the timing thereof; fluctuations in interest and currency exchange rates; claims or concerns regarding the safety or efficacy of marketed products or product candidates; the impact of health crises, like the novel coronavirus pandemic, on Takeda and its customers and suppliers, including foreign governments in countries in which Takeda operates, or on other facets of its business; the timing and impact of post-merger integration efforts with acquired companies; the ability to divest assets that are not core to Takeda's operations and the timing of any such divestment(s); and other factors identified in Takeda's most recent Annual Report on Form 20-F and Takeda's other reports filed with the U.S. Securities and Exchange Commission, available on Takeda's website at: https://www.takeda.com/investors/reports/sec-filings/ or at http://www.sec.gov. Takeda does not undertake to update any of the forward-looking statements contained in this press release or any other forward-looking statements it may make, except as required by law or stock exchange rule. Past performance is not an indicator of future results and the results or statements of Takeda in this press release may not be indicative of, and are not an estimate, forecast, guarantee or projection of Takeda's future results.

Continued here:
Takeda Pharmaceutical : China announces ADCETRIS (brentuximab vedotin) is approved for the treatment of adult patients with CD30-positive Lymphomas -...

New Data for Investigational CRISPR/Cas9 Gene-Editing Therapy CTX001 for Severe Hemoglobinopathies Accepted for Oral Presentation at the 25th European…

ZUG, Switzerland and CAMBRIDGE, Mass. and BOSTON, May 14, 2020 (GLOBE NEWSWIRE) -- CRISPR Therapeutics (Nasdaq: CRSP) and Vertex Pharmaceuticals Incorporated (Nasdaq: VRTX) today announced that new data from two ongoing Phase 1/2 clinical trials of the CRISPR/Cas9 gene-editing therapy CTX001 in severe hemoglobinopathies have been accepted for an oral presentation at the EHA Congress, which will take place virtually from June 11-14, 2020.

An abstract posted online today includes 12 months of follow-up data for the first patient treated in the ongoing Phase 1/2 CLIMB-111 trial in transfusion-dependent beta thalassemia (TDT) and 6 months of follow-up data for the first patient treated in the ongoing Phase 1/2 CLIMB-121 trial in severe sickle cell disease (SCD). Updated data will be presented at EHA, including longer duration follow-up data for the first two patients treated in these trials and initial data for the second patient treated in the CLIMB-111 trial.

The accepted abstract is now available on the EHA conference website: https://ehaweb.org/congress/eha25/key-information-2/.

Abstract Title: Initial Safety and Efficacy Results With a Single Dose of Autologous CRISPR-Cas9 Modified CD34+ Hematopoietic Stem and Progenitor Cells in Transfusion-Dependent -Thalassemia and Sickle Cell DiseaseSession Title: Immunotherapy - ClinicalAbstract Code: S280

About the Phase 1/2 Study in Transfusion-Dependent Beta ThalassemiaThe ongoing Phase 1/2 open-label trial, CLIMB-Thal-111, is designed to assess the safety and efficacy of a single dose of CTX001 in patients ages 18 to 35 with TDT. The study will enroll up to 45 patients and follow patients for approximately two years after infusion. Each patient will be asked to participate in a long-term follow-up study.

About the Phase 1/2 Study in Sickle Cell DiseaseThe ongoing Phase 1/2 open-label trial, CLIMB-SCD-121, is designed to assess the safety and efficacy of a single dose of CTX001 in patients ages 18 to 35 with severe SCD. The study will enroll up to 45 patients and follow patients for approximately two years after infusion. Each patient will be asked to participate in a long-term follow-up study.

About CTX001CTX001 is an investigational ex vivo CRISPR gene-edited therapy that is being evaluated for patients suffering from TDT or severe SCD in which a patients hematopoietic stem cells are engineered to produce high levels of fetal hemoglobin (HbF; hemoglobin F) in red blood cells. HbF is a form of the oxygen-carrying hemoglobin that is naturally present at birth and is then replaced by the adult form of hemoglobin. The elevation of HbF by CTX001 has the potential to alleviate transfusion requirements for TDT patients and painful and debilitating sickle crises for SCD patients. CTX001 is the most advanced gene-editing approach in development for beta thalassemia and SCD.

CTX001 is being developed under a co-development and co-commercialization agreement between CRISPR Therapeutics and Vertex.

About the CRISPR-Vertex CollaborationCRISPR Therapeutics and Vertex entered into a strategic research collaboration in 2015 focused on the use of CRISPR/Cas9 to discover and develop potential new treatments aimed at the underlying genetic causes of human disease. CTX001 represents the first treatment to emerge from the joint research program. CRISPR Therapeutics and Vertex will jointly develop and commercialize CTX001 and equally share all research and development costs and profits worldwide.

About CRISPR TherapeuticsCRISPR Therapeutics is a leading gene editing company focused on developing transformative gene-based medicines for serious diseases using its proprietary CRISPR/Cas9 platform. CRISPR/Cas9 is a revolutionary gene editing technology that allows for precise, directed changes to genomic DNA. CRISPR Therapeutics has established a portfolio of therapeutic programs across a broad range of disease areas including hemoglobinopathies, oncology, regenerative medicine and rare diseases. To accelerate and expand its efforts, CRISPR Therapeutics has established strategic partnerships with leading companies including Bayer, Vertex Pharmaceuticals and ViaCyte, Inc. CRISPR Therapeutics AG is headquartered in Zug, Switzerland, with its wholly-owned U.S. subsidiary, CRISPR Therapeutics, Inc., and R&D operations based in Cambridge, Massachusetts, and business offices in San Francisco, California and London, United Kingdom. For more information, please visit http://www.crisprtx.com.

CRISPR Forward-Looking StatementThis press release may contain a number of forward-looking statements within the meaning of the Private Securities Litigation Reform Act of 1995, as amended, including statements regarding CRISPR Therapeutics expectations about any or all of the following: (i) the status of clinical trials (including, without limitation, the expected timing of data releases) related to product candidates under development by CRISPR Therapeutics and its collaborators, including expectations regarding the data that is expected to be presented at the European Hematology Associations upcoming congress; (ii) the expected benefits of CRISPR Therapeutics collaborations; and (iii) the therapeutic value, development, and commercial potential of CRISPR/Cas9 gene editing technologies and therapies. Without limiting the foregoing, the words believes, anticipates, plans, expects and similar expressions are intended to identify forward-looking statements. You are cautioned that forward-looking statements are inherently uncertain. Although CRISPR Therapeutics believes that such statements are based on reasonable assumptions within the bounds of its knowledge of its business and operations, forward-looking statements are neither promises nor guarantees and they are necessarily subject to a high degree of uncertainty and risk. Actual performance and results may differ materially from those projected or suggested in the forward-looking statements due to various risks and uncertainties. These risks and uncertainties include, among others: the potential impacts due to the coronavirus pandemic, such as the timing and progress of clinical trials; the potential for initial and preliminary data from any clinical trial and initial data from a limited number of patients (as is the case with CTX001 at this time) not to be indicative of final trial results; the potential that CTX001 clinical trial results may not be favorable; that future competitive or other market factors may adversely affect the commercial potential for CTX001; uncertainties regarding the intellectual property protection for CRISPR Therapeutics technology and intellectual property belonging to third parties, and the outcome of proceedings (such as an interference, an opposition or a similar proceeding) involving all or any portion of such intellectual property; and those risks and uncertainties described under the heading "Risk Factors" in CRISPR Therapeutics most recent annual report on Form 10-K, and in any other subsequent filings made by CRISPR Therapeutics with the U.S. Securities and Exchange Commission, which are available on the SEC's website at http://www.sec.gov. Existing and prospective investors are cautioned not to place undue reliance on these forward-looking statements, which speak only as of the date they are made. CRISPR Therapeutics disclaims any obligation or undertaking to update or revise any forward-looking statements contained in this press release, other than to the extent required by law.

About VertexVertex is a global biotechnology company that invests in scientific innovation to create transformative medicines for people with serious diseases. The company has multiple approved medicines that treat the underlying cause of cystic fibrosis (CF) a rare, life-threatening genetic disease and has several ongoing clinical and research programs in CF. Beyond CF, Vertex has a robust pipeline of investigational small molecule medicines in other serious diseases where it has deep insight into causal human biology, including pain, alpha-1 antitrypsin deficiency and APOL1-mediated kidney diseases. In addition, Vertex has a rapidly expanding pipeline of genetic and cell therapies for diseases such as sickle cell disease, beta thalassemia, Duchenne muscular dystrophy and type 1 diabetes mellitus.

Founded in 1989 in Cambridge, Mass., Vertex's global headquarters is now located in Boston's Innovation District and its international headquarters is in London, UK. Additionally, the company has research and development sites and commercial offices in North America, Europe, Australia and Latin America. Vertex is consistently recognized as one of the industry's top places to work, including 10 consecutive years on Science magazine's Top Employers list and top five on the 2019 Best Employers for Diversity list by Forbes. For company updates and to learn more about Vertex's history of innovation, visit http://www.vrtx.com/ or follow us on Facebook, Twitter, LinkedIn, YouTube and Instagram.

Vertex Special Note Regarding Forward-Looking StatementsThis press release contains forward-looking statements as defined in the Private Securities Litigation Reform Act of 1995, including, without limitation, information regarding the data that is expected to be presented at the European Hematology Association (EHA)s upcoming Congress. While Vertex believes the forward-looking statements contained in this press release are accurate, these forward-looking statements represent the company's beliefs only as of the date of this press release and there are a number of factors that could cause actual events or results to differ materially from those indicated by such forward-looking statements. Those risks and uncertainties include, among other things, that the development of CTX001 may not proceed or support registration due to safety, efficacy or other reasons, and other risks listed under Risk Factors in Vertex's annual report and quarterly reports filed with theSecurities and Exchange Commissionand available through the company's website atwww.vrtx.com. Vertex disclaims any obligation to update the information contained in this press release as new information becomes available.

(VRTX-GEN)

CRISPR Therapeutics Investor Contact:Susan Kim, +1 617-307-7503susan.kim@crisprtx.com

CRISPR Therapeutics Media Contact:Rachel EidesWCG on behalf of CRISPR+1 617-337-4167 reides@wcgworld.com

Vertex Pharmaceuticals IncorporatedInvestors:Michael Partridge, +1 617-341-6108orZach Barber, +1 617-341-6470orBrenda Eustace, +1 617-341-6187

Media:mediainfo@vrtx.com orU.S.: +1 617-341-6992orHeather Nichols: +1 617-839-3607orInternational: +44 20 3204 5275

See the rest here:
New Data for Investigational CRISPR/Cas9 Gene-Editing Therapy CTX001 for Severe Hemoglobinopathies Accepted for Oral Presentation at the 25th European...

Dlp-mediated Hh and Wnt signaling interdependence is critical in the niche for germline stem cell progeny differentiation – Science Advances

INTRODUCTION

Stem cells in adult tissue undergo lifelong continuous self-renewal and generate differentiated cells for maintaining tissue homeostasis by replenishing the lost cells caused by natural turnover, aging, injury, or disease. Adult stem cell self-renewal and proliferation are demonstrated to be controlled by the niche in various tissues and organisms (1, 2). Studies on stem cells from different organisms ranging from Drosophila to mammals have demonstrated that one or multiple signals originated from the niche directly act on stem cells in concert with varieties of different intrinsic factors to control stem cell self-renewal by repressing differentiation pathways (1, 2). Our recent study on germline stem cells (GSCs) in the Drosophila ovary has also demonstrated that stem cell progeny differentiation is also controlled extrinsically by the niche formed by adjacent stromal cells, which is named as the differentiation niche (3). Resident macrophage cells on the surface seminiferous tubule in the adult mouse testis also contribute to the niche for regulating germ cell differentiation, suggesting that the niche dedicated to differentiation might also exist in mammalian tissues (4). Multiple signaling pathways are usually used by niches in various stem cell systems to control either self-renewal or differentiation, but how they cooperate with one another in niches to control stem cell behavior remains poorly understood. In this study, we show that Hh and Wnt signaling pathways use novel cooperative mechanisms to provide the favorable environment for GSC progeny differentiation in the Drosophila ovary by maintaining each others signaling activities.

The Drosophila ovary provides an effective system for studying stem cell self-renewal and differentiation due to well-defined GSCs and niches. Two or three GSCs physically interact with the niche consisting of primarily cap cells, whereas early GSC progeny physically interact with their own niche composed of inner germarial sheath (IGS) cells (also known as escort cells) (fig. S1A) (5, 6). The GSCs located at the tip of the germarium continuously generate cystoblasts (CBs), which can further divide four times synchronously with incomplete cytokinesis to form mitotic cysts (2-cell, 4-cell, and 8-cell) and 16-cell cysts. Cap cells and anterior row of IGS cells directly contact GSCs and form the niche for promoting self-renewal (710). The niche uses bone morphogenetic protein (BMP) signaling and E-cadherinmediated cell adhesion to control GSC self-renewal and proliferation (5). In 2011, IGS cells were first proposed to form a niche for promoting GSC progeny differentiation (3). Thus, two distinct niche compartments control stem cell self-renewal and differentiation separately.

IGS cells function as the differentiation niche for GSC progeny through physical interactions and signaling. IGS cells extend long cellular processes to encase early differentiating GSC progeny, including CBs, mitotic cysts, and 16-cell cysts (3, 11). Further genetic studies have demonstrated that IGS cellular processmediated physical interactions are essential for promoting GSC progeny differentiation (3, 9, 12, 13). In addition, different signaling pathways and gene networks have been identified for their requirement in IGS cells for promoting GSC progeny differentiation by preventing BMP signaling through distinct mechanisms (5). Notably, epidermal growth factor receptor (EGFR), Wnt, Hh, and Jak-Stat signaling pathways are required in IGS cells to promote GSC progeny differentiation by preventing BMP signaling via regulation of dally, dpp, or both (12, 1421). dally encodes a proteoglycan protein promoting the diffusion of Dpp/BMP protein in Drosophila (22). In addition to repressing BMP signaling, IGS cells might also send direct signals to GSC progeny to promote their differentiation, but these signals remain to be defined. In contrast, EGFR signaling has so far been reported to be required in adult somatic cysts to promote GSC progeny differentiation in the Drosophila testis (23).

Since both Hh and Wnt signaling pathways are required in IGS cells for promoting GSC progeny differentiation by repressing BMP signaling, their functional relationship in the niche remains unclear. This study has revealed that they are dependent on each other for their activities in IGS cells through repressing dally-like protein (dlp). dlp encodes a Dally-related glypican (GPC) protein, which is known to promote BMP, Hh, and Wnt signaling in Drosophila (24). However, Dlp-related GPCs can both promote and inhibit Shh and Wnt signaling in mammals (2527). Here, we show that Dlp up-regulation can sufficiently inhibit both Hh and Wnt signaling and elevate BMP signaling. dlp knockdown in IGS cells can significantly rescue the GSC progeny differentiation defects caused by defective Hh or Wnt signaling and can also uncouple the interdependence of Hh and Wnt signaling. Hh and Wnt signaling directly repress dlp expression through recruiting Croc and H3K9 trimethylase Eggless into the regulatory region. Therefore, this study has revealed a novel cooperative mechanism of Hh and Wnt signaling and a novel Hh/Wnt-mediated mechanism for dlp repression in the niche for preventing BMP signaling and promoting GSC progeny differentiation.

Hh and Wnt signaling are both required in IGS cells for proper GSC progeny differentiation. To investigate the relationship between Hh and Wnt signaling in IGS cells, we examined the expression of ptcgreen fluorescent protein (GFP) and fz3red fluorescent protein (RFP), which are Hh and Wnt signaling activity reporters, respectively (2830), in adult smo and dsh knockdown (smoKD and dshKD) IGS cells. IGS-expressed gal4 line, c587, is combined with a temperature-sensitive mutant gal80 (gal80ts) to achieve RNA interference (RNAi)mediated gene knockdown in adult IGS cells after shifting adult flies from room temperature to 29C (fig. S1B) (12, 15). Two independent RNAi lines for smo and dsh, which had been characterized previously (12, 15), were used to inactivate Hh or Wnt signaling in this study, respectively. The enhancer trap line PZ1444 expressing nuclear LacZ is used to label IGS cells and cap cells, which can further be distinguished on the basis of their distinct nucleus size and location (15). Consistent with our previous finding that Hh and Wnt signaling are required for IGS maintenance, most of IGS cells are lost 5 and 7 days after dsh or smo knockdown (fig. S1, C and D). However, IGS numbers remain close to normal 2 days after their knockdown, which is the time when we examined fz3-RFP and ptc-GFP expression throughout this study (fig. S1, C and D, and Fig. 1, A and B).

The germaria are labeled for PZ1444-LacZ expression to visualize IGS cells (two indicated by arrowheads) and cap cells (broken ovals), while DAPI staining identifies all nuclei. (A to D) Merged confocal images of germaria showing that the expression of both fz3-RFP (A) and ptc-GFP (B) is significantly decreased in smoKD and dshKD IGS cells 2 days after knocking down compared with the control (lucKD) (C and D) quantification results on fz3-RFP or ptc-GFP intensities normalized to LacZ in IGS cells, respectively; n = IGS cells number. (E to H) Merged FISH (green) and immunostaining (LacZ, red) confocal images showing that fz3 (E) or ptc (F) mRNA expression levels are significantly reduced in dshKD and smoKD IGS cells (G and H: quantification results on fz3 and ptc mRNA levels based on the fluorescence intensities normalized to LacZ, respectively; n = germarial number). Scale bars, 10 m (all images at the same scale). In this study, all the quantitative data are shown as means SEM, whereas P values are determined by the two-sided Students t test (***P 0.001; **P 0.01).

On the basis of fz3-RFP and ptc-GFP expression, knocking down smo or dsh for 2 days can effectively inactivate Hh and Wnt signaling in adult IGS cells, respectively (Fig. 1, A to D). Adult dshKD IGS cells significantly decrease ptc-GFP expression, while smoKD IGS cells significantly reduce fz3-RFP expression, indicating that Hh and Wnt signaling regulate each other (Fig. 1, A to D). Our previous RNA sequencing (RNA-seq) results on purified dshKD IGS cells did not show significant changes in fz3 and ptc mRNAs compared with control IGS cells (table S1) (15). One of the concerns is that enzymatic dissociation of IGS cells destroys Hh and Wnt proteins in the extracellular space, which result in the loss of Hh and Wnt signaling in control IGS cells. As the whole IGS purification process lasts about 4 hours, which might be long enough for fz3 and ptc mRNA decay, fluorescence-activated cell sorting (FACS)purified control and dshKD IGS cells behave similarly on fz3 and ptc mRNA levels. In the future, it should be extremely cautious to use FACS-purified cells for examining gene expression changes caused by secreted factors. Then, we performed fluorescent mRNA in situ hybridization (FISH) using quantitative hybridization chain reaction technology to further examine fz3 and ptc mRNA expression changes in dshKD or smoKD IGS cells (31). dshKD or smoKD significantly decreases the expression of both fz3 and ptc mRNAs in IGS cells (Fig. 1, E to H). To exclude the possibility that germ cell defects cause the loss of fz3-RFP and ptc-GFP expression in IGS cells, we examine fz3-RFP and ptc-GFP expression in IGS-specific tkv knockdown germaria, which exhibit the severe germ cell differentiation defect as reported previously (fig. S1, E and F) (18, 32). fz3-RFP and ptc-GFP expression remain normal in tkvKD IGS cells despite the presence of the severe differentiation defect (fig. S1, E and F). Together, these results indicate that Wnt and Hh signaling are mutually dependent in IGS cells.

To investigate the mechanism underlying the Hh and Wnt signaling interdependence, we examined the previous RNA-seq results on purified control and dshKD IGS cells (15). dlp mRNA is up-regulated by about fourfold, but dally expression remains unchanged, in dshKD IGS cells compared with control ones (fig. S2A and table S1). dally and dlp encode highly related glypican proteins known to modulate Dpp/BMP, Hh, and Wnt signaling in Drosophila (24). Although its mRNA and protein levels are very low in control IGS cells, dlp mRNA and protein levels are drastically up-regulated in dshKD and smoKD IGS cells based on FISH and immunostaining results, respectively (fig. S2, B and C, and Fig. 2, A and B). These results reveal that Hh and Wnt signaling are required in IGS cells to repress Dlp expression.

The germaria (A, E, and F) are labeled for PZ1444-LacZ to visualize IGS cells (two by arrowhead) and cap cells (broken ovals), while DAPI staining identifies all nuclei. (A) Merged confocal images of germaria showing that Dlp protein (green) levels are significantly up-regulated in the dshKD1 and smoKD1 IGS cells 2 days after knockdown (B) quantification results normalized to LacZ; n = germarial number. (C) dlp overexpression (dlpOE) in IGS cells causes the accumulation of significantly more spectrosome-containing undifferentiated SGCs (only two in control and four in dlpOE indicated by arrows) in 5- and 10-day-old germaria but have no or a little impact on GSCs (highlighted by broken ovals) (D) CB/SGC and GSC quantification results; n = germarial number. (E to H) ptc-GFP and fz3-RFP expression is drastically down-regulated in dlpOE IGS cells (arrowheads) 2 days after overexpression compared with the control (G and H: quantification results on fz3-RFP or ptc-GFP intensities normalized to LacZ in each IGS cell; n = germarial number). (I to K) Merged confocal images showing that dlpOE germaria accumulate more bam-GFPnegative, pMad-positive, and Dad-LacZpositive SGCs (some by arrows in J and K). (L to N) Inactivating one copy of dpp by dpphr27/+ or dpphr56 significantly decreases both pMad-positive SGCs (L) and GSC accumulation (M) caused by dlpOE without any obvious effect on GSC numbers (N: CB/SGC and GSC quantification results; n = germarial number). Scale bars, 10 m. (***P 0.001; *P 0.05).

Then, we determined whether dlp up-regulation in IGS cells can affect GSC progeny differentiation, Hh and Wnt signaling. In contrast with the control germaria containing about one CB, the 5- and 10-day Dlp-overexpressing (dlpOE) germaria accumulate approximately 10 and 20 spectrosome-containing single germ cells (SGCs), respectively, indicating that Dlp overexpression blocks CB differentiation (Fig. 2, C and D). dlp overexpression also diminishes the expression of fz3-RFP and ptc-GFP in IGS cells, indicating that Dlp up-regulation can sufficiently repress both Hh and Wnt signaling activities in the niche (Fig. 2, E to H). Dlp is known to be directly associated with Dpp, Hh, and Wg proteins to modulate their signaling activity (22, 26, 33, 34), but it remains undermined whether Dlp can also directly bind to Wnt2 and Wnt4, two highly expressed Wnt proteins in IGS cells (15). We used coimmunoprecipitation (co-IP) experiments in S2 cells to show that Dlp can also be associated with Wnt2 and Wnt4 (fig. S2, D to G). Together, our findings indicate that up-regulated Dlp expression sufficiently and directly represses Hh and Wnt signaling in IGS cells.

BMP signaling elevation is known to be linked to the CB differentiation defects caused by defective Hh and Wnt signaling in IGS cells (12, 1518). In control germaria, BMP signaling leads to production of phosphorylated Mad (pMad), activation of Dad-lacZ reporter expression in GSCs, and represses bam-GFP (Fig. 2, I to K). In GSC progeny, including CBs, pMad and Dad-lacZ are turned off and bam-GFP is activated due to the absence of BMP signaling (Fig. 2, I to K). However, in the niche-specific Dlp-overexpressing germaria, most of the accumulated SGCs are positive for pMad and Dad-lacZ but negative for bam-GFP, indicating that Dlp overexpression in IGS cells elevates BMP signaling in GSC progeny, thus blocking their differentiation (Fig. 2, I to K). dpphr56 and dpphr27 heterozygous mutations were used previously to effectively decrease BMP signaling in the Drosophila ovary because dpp encodes a major BMP ligand in the ovary (12, 15, 35). Compared with dlpOE, the removal of one copy of dpp significantly reduces pMad level in germarium (Fig. 2L). dpphr56 and dpphr27 heterozygous mutations can significantly rescue the CB differentiation defects caused by dlp overexpression, but do not affect GSC numbers significantly (Fig. 2, M and N). Therefore, our experimental results demonstrate that Dlp up-regulation in IGS cells increases BMP signaling, thereby disrupting GSC progeny differentiation.

Since Dlp is known to regulate Hh and Wnt signaling in Drosophila (36, 37), we then determined if Dlp is required for modulating Hh and Wnt signaling in IGS cells. Although two dlp short hairpin RNA (shRNA) lines can efficiently knock down Dlp protein expression (fig. S3, A to D), dlp knockdown does not affect fz3-RFP and ptc-GFP expression in IGS cells, indicating that endogenous Dlp is dispensable for Hh and Wnt signaling activities in the differentiation niche (fig. S3, E and F). dlp knockdown in IGS cells can significantly rescue fz3-GFP expression in the smoKD IGS cells as well as ptc-GFP expression in the smoKD IGS cells (Fig. 3, A to C). These results indicate that Dlp repression is required for maintaining Hh and Wnt signaling independence in IGS cells.

(A and B) dlp knockdown in IGS cells can significantly and drastically rescue the expression of ptc-GFP and fz3-RFP in dshKD and smoKD IGS cells (arrowheads), respectively (broken ovals highlight cap cells; C) quantification results on ptc-GFP and fz3-RFP fluorescence intensities normalized to LacZ; n = IGS cell numbers. (D and E) dlp knockdown in IGS cells can significantly decrease the accumulation of SGCs (only some of them by arrowheads) caused by dshKD and smoKD (E: CB/SGC quantification results; n = germarial number). Scale bars, 10 m. (***P 0.001; **P 0.01).

To determine whether dlp up-regulation is responsible for the germ cell differentiation defects caused by defective Hh and Wnt signaling, we examined the SGC accumulation in dlpKD dshKD and dlpKD smoKD germaria. The dlpKD lucKD germaria contain similar GSC and CB numbers to those lucKD germaria, indicating that Dlp is also dispensable in IGS cells for promoting GSC progeny differentiation (Fig. 3, D and E). As expected, dlp knockdown in IGS cells can significantly decrease the Dlp up-regulation in IGS cells and reduce the SGC accumulation caused by smoKD or dshKD but cannot rescue the germ cell differentiation defects completely, indicating that Hh and Wnt signaling promote GSC progeny differentiation partly by repressing dlp expression (Fig. 3, D and E, and fig. S3, A to D). Reducing the dlp dosage by three independent heterozygous mutations can significantly decrease Dlp protein expression in IGS cells and also rescue the germ cell differentiation defects caused by defective Hh and Wnt signaling in IGS cells (fig. S4, A to E). This partial rescue by dlpKD and heterozygous dlp mutations can be explained by the previous findings that Hh and Wnt signaling have additional important downstream targets in addition to dlp (12, 15, 20). dlpKD can only decrease the IGS cell loss caused by dshKD or smoKD 3 days, but not 5 days, after knockdown, suggesting that Hh and Wnt signaling maintain IGS cells largely independent of Dlp repression (fig. S4, F to G). Therefore, these results show that Hh and Wnt signalingmediated dlp repression in IGS cells is required for their signaling interdependence and normal GSC progeny differentiation.

To further investigate how Wnt and Hh signaling repress dlp expression in IGS cells, we generated a series of transgenes carrying different dlp genomic fragments and followed with the GFP complementary DNA (cDNA) by using the pGreenRabbit (pGR) vector (Fig. 4A)(38). Through two rounds of genomic fragment screens, a 900base pair (bp) genomic region (dlp2.1.5) in the second intron is identified to sufficiently recapitulate Dlp expression patterns in the germarium (Fig. 4B and fig. S5A). Both dlp2.1.5-GFP and endogenous Dlp protein show low expression in IGS cells and high expression in follicle cells (Fig. 4B). Consistent with the idea that the dlp2.1.5 genomic region carries most, if not all, of regulatory elements for Hh/Wnt signalingmediated repression, dlp2.1.5-GFP is up-regulated in both smoKD and dshKD IGS cells (Fig. 4C and fig. S5B). Transcription factors Ci and Pan function downstream of Hh and Wnt signaling, respectively, to regulate target gene expression (3941). dlp2.1.5-GFP is also up-regulated in ciKD and panKD IGS cells, indicating that canonical Hh and Wnt signaling likely repress dlp in IGS cells via the 900-bp region (Fig. 4C and fig. S5B). dlp overexpression can also up-regulate dlp2.1.5-GFP expression, suggesting that there is a feedforward loop via Hh/Wnt signaling for the dlp control. These results indicate that Hh and Wnt signaling repress dlp transcription via a small regulatory region.

(A) Diagram of the dlp genomic regions showing dlp2.1 and dlp2.1.5 regions driving GFP expression, which recapitulates dlp mRNA and protein expression in the germarium (please see fig. S5 for details). (B) Immunostaining with anti-GFP (green) and anti-Dlp (red) showing dlp2.1.5-GFP has similar expression pattern with endogenous Dlp, which has very low level at IGS cells but high level at late-stage somatic cells. (C) Single confocal cross-sectional images of germaria showing that dlp2.1.5-GFP expression is up-regulated in dshKD1, smoKD1, panKD, ciKD, and dlpOE IGS cells compared with the control (lucKD) (anterior germarial regions highlighted by squares are shown at a high magnification). Scale bars, 20 m in (B), 10 m in (C).

To further define individual elements in the dlp2.1.5 region for Hh/Wnt signalingmediated repression in IGS cells, we generated GFP reporter transgenic flies carrying nested deletions from both the ends of the 900-bp dlp2.1.5 region with each deleting 100 bp, dlp2.1.5 1-GFP to dlp2.1.5 10-GFP (fig. S6A). Our nested deletion analyses in wild-type, dshKD, and smoKD IGS cells have yielded three pieces of important information. First, only an 800-bp continuous genomic region is sufficient for recapitulating dlp expression in the germarium since dlp2.1.5 6-GFP has the same expression patterns and levels as dlp2.1.5-GFP (fig. S6H). Second, multiple repressive elements likely scatter along the 800-bp genomic region to work synergistically for Hh/Wnt signalingmediated dlp repression in IGS cells since no single 100-bp deletion in the 800-bp region alone sufficiently causes the up-regulation of the GFP reporter in IGS cells (fig. S6, B to G and I to L). Third, multiple activators in the 800-bp region are required for Dlp gene expression in follicle cells and also for defective Hh/Wnt signalingcaused Dlp up-regulation in IGS cells. On the basis of GFP expression in follicle cells, the deleted regions in dlp2.1.5 1, dlp2.1.5 5, dlp2.1.5 7, and dlp2.1.5 8 are important for dlp expression in follicle cells and equally important for dshKD/smoKD-mediated dlp up-regulation. Among them, the deleted regions by dlp2.1.5 1 and dlp2.1.5 8 have stronger effects on dlp gene activation in follicle cells and also have stronger suppression effects on defective Hh/Wnt signalingmediated dlp up-regulation than those deleted ones in dlp2.1.5 5 and dlp2.1.5 7, suggesting that scattered repressive elements in the 800-bp region suppress dlp expression in IGS cells likely by antagonizing the activators. To determine whether the 800-bp-long regulatory region is required for endogenous Dlp protein expression in follicle cells, we used CRISPR-Cas9 to generate a dlp215 mutant deleting the region, which homozygotes are lethal likely due to its requirement for Dlp expression during early development (fig. S6N). In the dlp215 heterozygous mutant germarium, Dlp protein expression is deceased in follicle cells as predicted (fig. S6, O and P). Together, these results suggest that Hh/Wnt signalingmediated dlp repression in the niche is accomplished through multiple cooperative repressive elements in the dlp regulatory region.

Then, we performed the electrophoretic mobility shift assay (EMSA) to determine whether Ci and Pan directly bind to multiple sites in the 800-bp region using overlapped biotin-labeled 24-bp DNA fragments and purified glutathione S-transferase (GST) fusion proteins with Ci and Pan DNA binding domains (Fig. 5Aand fig. S7A). Our EMSA assay has identified four strong Ci binding regions and three strong Pan binding regions in the 800-bp genomic region in addition to some weak binding sites (Fig. 5A). Two of the four Ci binding sites, CGTGGCTGGC and GACAAGGGACT, are consistent with bioinformatic prediction, whereas only one of the three Pan binding sites, GGATACCAAAAATAGG, is predicted, suggesting that Ci and Pan are capable of binding to the previously uncharacterized new sites. Chromatin immunoprecipitation (ChIP) results have further confirmed that Ci and Pan also associate with dlp2.1.5 region in vivo and show stronger enrichment near in vitroidentified binding regions (Fig. 5, B and C, and fig. S7B).

(A) EMSA results showing that GST-Ci-ZNF binds to four sites strongly and additional few sites weakly (green), while GST-Pan-HMG binds to three sites strongly and one site weakly (blue) (* and ** indicate weak and strong sites, respectively). (B) ChIP-qPCR results show that IGS-expressed Flag-Ci or Flag-Pan is associated in vivo with the 800 bp of dlp2.1.5 (P values compared with background control act5C). Mouse IgG antibodies were used as a negative IP control. (***P 0.001). (C) Summary diagram displaying Pan and Ci binding sites/regions in the dlp2.1.5 region based on EMSA and ChIP results (A and B), as well as the expression-activating regions based on the deletion results in fig. S6. (D) Mutating all the strong binding sites for Pan, Ci, or both causes a moderate GFP up-regulation in IGS cells compared with dlp2.1.5-GFP (note: since these mutations also decrease dlp2.1.5-GFP expression in follicle cells, GFP expression in follicle cells are normalized to that in normal dlp2.1.5-GFP). Scale bars, 10 m.

These Ci and Pan binding regions overlap with the activating regions, suggesting that Ci and Pan binding to the regulatory region might preclude the binding of currently uncharacterized transcription activators (Fig. 5C). Diminishing the binding activities of Ci, Pan, or both in dlp2.1.5 by mutating the experimentally defined sites results in up-regulated GFP reporter expression in IGS cells, showing that Wnt and Hh signaling directly repress dlp expression in IGS cells (Fig. 5D and fig. S7, C and D). It is worth noting that dlp2.1.5-GFP reporter up-regulation caused by the mutated Ci or Pan binding sites is relative moderate compared with that caused by defective Hh and Wnt signaling, suggesting that Ci and Pan binding sites might overlap with the activators binding sites in the identified dlp regulatory region. Together, Hh and Wnt pathway downstream transcription factors Ci and Pan can directly bind to multiple sites in the dlp regulatory region to repress dlp expression in the niche.

To further investigate how Hh and Wnt signaling repress dlp transcription via the 800-bp genomic region in IGS cells, we then investigated how it works with Wnt signaling in IGS cells to directly repress dlp expression by carrying out the shRNA knockdown of transcription factors expressed in IGS cells. In the screen, crocodile (croc) and eggless (egg) were identified for their requirement in repressing dlp2.1.5-GFP expression in IGS cells. Knockdown of croc or egg results in the up-regulated expression of dlp2.1.5-GFP compared with the control (Fig. 6A). Consistent with this, crocKD or eggKD IGS cells also show increased Dlp protein expression (fig. S8, A and B). egg encodes an H3K9 trimethylase, which has been shown previously to be required in IGS cells for promoting GSC progeny differentiation (9, 42). croc encodes a fork head domaincontaining transcriptional factor (43). Consistent with the idea that Egg and Croc are involved in Dlp repression in IGS cells, knocking down egg or croc in IGS cells also leads to a significant down-regulation of Wnt and Hh signaling (fig. S8, C to F). These results suggest that Egg and Croc might be involved in Hh/Wnt signalingmediated dlp repression in IGS cells.

(A) Compared with WT, dlp2.1.5-GFP expression is up-regulated in eggKD and crocKD IGS cells (brackets) 5 days after knockdown. (B and C) Fourteen-day croc knockdown in IGS cells causes the accumulation of SGCs (arrowheads) (C: CB/SGC and GSC quantification results). n.s., no significance. (D) Summary diagram showing the Croc binding sites in the dlp2.1.5 region based on EMSA and ChIP results in fig. S8 (H to J). (E) ChIP-qPCR results show that the binding ability of Croc to dlp2.1.5 is significantly reduced in smoKD or dshKD IGS cells compared with WT. (F and G) In S2 cells, CiPKA-Myc and Pan-HA can bring down Croc-Flag. CiPKA is a noncleavable active full-length Ci, whereas ArmS10-Myc is the active Arm protein, which can bind to Pan for nuclear import in the absence of Wnt signaling. (H) In S2 cells, Croc-HA can bring down Croc-Flag, indicative of potential dimerization or oligomerization. (I) In S2 cells, Croc-HA can bring down Egg-Flag (IgG as a negative control: *, a nonspecific protein recognized by the anti-HA antibody). (J and K) Coexpression of Croc-HA can significantly increase Egg-Flag protein levels in S2 cells (empty plasmid used to normalize total transfected DNA; K: quantification results on Egg-Flag and Croc-HA levels). (L) Schematic diagram explaining how Hh/Wnt signalingmediated direct Dlp repression maintains their interdependence and prevents BMP signaling, thereby promoting GSC progeny differentiation. Scale bars, 10 m. (***P 0.001).

Then, we determined whether Croc is also required in IGS cells to promote GSC progeny differentiation by directly repressing dlp expression. Knocking down croc in IGS cells by two independent shRNAs results in the accumulation of SGCs but does not affect GSC maintenance, indicating that Croc is required in the niche to promote GSC progeny differentiation (Fig. 6, B and C). In addition, our EMSA results indicate that Croc protein can also bind to two sites in the 800-bp dlp regulatory region (Fig. 6D and fig. S8, G and H). ChIPquantitative polymerase chain reaction (qPCR) results have further shown that IGSspecifically expressed Croc-HA binds strongly in vivo to the in vitroidentified binding sites in the dlp2.1.5 region (fig. S8, I and J). Notably, the Croc in vivo binding ability to the dlp regulatory region is significantly decreased in the smoKD and dshKD IGS cells compared with the control (Fig. 6E). Together, these results reveal that Croc binds to the dlp regulatory region to repress dlp expression in IGS cells, thereby promoting GSC progeny differentiation.

To further investigate how they work together to repress dlp in IGS cells, we tested whether Croc, Ci, and Pan can associate with each other in S2 cells. Myc-tagged stable full-length CiPKA-Myc, which is PKA phosphorylation resistant for preventing its cleavage (44), can pull down Flag-tagged Croc (Flag-Croc) (Fig. 6F). Similarly, hemagglutinin (HA)tagged Pan (Pan-HA) can also bring down Croc-Flag in the presence of Wnt signaling activated ArmS10, which forms a protein complex with Pan for its nuclear import (Fig. 6G)(45). HA-tagged Croc (Croc-HA) can also precipitate Croc-Flag, indicating that Croc proteins can dimerize or oligomerize (Fig. 6H). Flag-tagged Egg (Egg-Flag) could also be coimmunoprecipitated by Croc-HA (Fig. 6I). Croc can also significantly stabilize Egg-Flag in S2 cells in a dosage-dependent manner (Fig. 6, J and K). In summary, these results indicate that signaling-activated nuclear-localized Pan and Ci recruit Croc and, subsequently, Egg for promoting H3K9 trimethylation and, thus, blocking the access of the transcriptional activators to the dlp2.1.5 region and preventing dlp transcription in IGS cells.

Although Shh and Wnt signaling work synergistically to promote neural stem cell proliferation/differentiation in the mouse developing brain as well as cell proliferation in human medulloblastoma, the underlying mechanisms remain missing (46, 47). In addition, both Hh and Wnt signaling have also been shown to be required in the niche to promote GSC progeny differentiation in the Drosophila ovary by repressing BMP signaling, but the cooperative mechanisms are also not understood as well (15, 1719). In this study, we show that Hh and Wnt signaling sustain each other in the niche by directly repressing Dlp expression through Ci/Pan-recruited transcription factor Croc and H3K9 trimethylase Egg, and such repression is critical for preventing BMP signaling and, thus, promoting GSC progeny differentiations (Fig. 6L).

The cooperative and antagonistic relationships between Hh and Wnt signaling have been well established in normal developmental contexts and various human tumors. The antagonistic relationship between Hh and Wnt signaling is often accomplished through intrinsic signal transducers, target genes, and secreted inhibitors (48, 49). In Drosophila, Wg and Hh often regulate the same developmental processes synergistically through regulating each others expression (50, 51). However, the molecular mechanisms underlying Hh and Wnt signaling synergistic relationships remain largely unknown. Our findings have revealed a new Hh and Wnt signaling interdependent relationship maintained by a novel Dlp-mediated mechanism.

This study has provided the convincing experimental evidence supporting the Dlp repressionmediated Hh and Wnt signaling interdependence. First, Wnt and Hh signaling are required for each other to sustain their signaling activities in the niche. Second, Hh and Wnt signaling are required in the niche to directly repress Dlp expression since dlp mRNA and protein are significantly up-regulated in the Hh/Wnt signalingdefective niche. Third, niche-specific Dlp overexpression eliminates Hh/Wnt signaling in the niche. Fourth, Dlp overexpression can also further induce its own transcription, likely through inactivating Hh and Wnt signaling, suggesting that there is a feedforward loop for dlp regulation in the niche. Last, Dlp repression in the niche is critical for the interdependence of Hh and Wnt signaling. Together, our findings demonstrate that Hh/Wnt signalingmediated dlp repression is essential for maintaining the Hh/Wnt signaling interdependence in the niche (Fig. 6L). Although this study has revealed a novel mode for Hh/Wnt signaling cooperation as well as a novel mechanism mediating such cooperation, many important questions remain to be answered, such as how Dlp up-regulation inhibits Hh and Wnt signaling mechanistically in the niche and whether such regulatory mechanism operates in other developmental contexts and some diseased conditions.

BMP signaling activated by GSC niche-secreted Dpp is necessary and sufficient for maintaining GSC self-renewal by repressing differentiation (5). IGS cells function as a niche for promoting GSC progeny differentiation partly by preventing BMP signaling (3). Recent several studies have shown that Hh and Wnt signaling are required in IGS cells to promote GSC progeny differentiation partly by preventing BMP signaling via multiple mechanisms. Hh signaling functions in IGS cells to repress dpp expression and antagonize Hippo signaling, thereby preventing BMP signaling (12, 20). Wnt signaling is required in IGS cells to prevent BMP signaling activities in GSC by maintaining Tkv expression, IGS cellular processes, and the redox state, as well as by repressing dpp expression (1518). On the basis of our data, we propose a model that Hh and Wnt signaling function in IGS cells to prevent BMP signaling in GSC progeny by repressing dlp expression (Fig. 6L).

This study has provided several pieces of experimental evidence demonstrating that Hh/Wnt signalingmediated Dlp repression in IGS cells is essential for preventing BMP signaling and promoting GSC progeny differentiation. Dlp up-regulation in IGS cells causes BMP signaling elevation in GSC progeny as well as severe differentiation defects. In addition, decreasing BMP signaling by dpp mutations can significantly and drastically rescue the differentiation defects caused by Dlp overexpression in the niche. Furthermore, dlp knockdown in the niche can significantly rescue the GSC differentiation defects caused by defective Hh or Wnt signaling. Consistent with our findings, Dlp has been suggested to promote BMP signaling by increasing BMP concentration at the cell surface or functioning as a BMP coreceptor in Drosophila (24). It will be of great interest to investigate how Dlp mechanistically promotes BMP signaling in the differentiation niche.

Wnt signaling has been shown to directly repress the transcription of dpp in the leg imaginal disc by recruiting the Pan/Arm/Brinker complex to canonical T cell factor (TCF) binding sites and the transcription of Ugt36Bc in the hemocyte by recruiting the TCF/Pan complex to uncanonical TCF binding sites (52, 53). Hh signaling has only been reported to directly repress tkv expression in Drosophila wing imaginal disc via the full-length Ci, but the underlying mechanism remains unclear (54). This study shows that Hh and Wnt signaling can directly repress dlp expression by recruiting the Croc-Egg protein complex to TCF and Ci binding sites in the dlp regulatory region (Fig. 6L).

In this study, we have revealed that Hh and Wnt signaling downstream transcription factors Ci and Pan bind to multiple sites of a dlp regulatory region to antagonize activators, thereby repressing dlp expression. The 800-bp-long regulatory region in the second intron of dlp (dlp2.1.5) sufficiently recapitulates the expression pattern of Dlp protein in the Drosophila ovary based on the GFP reporters containing different dlp genomic fragments. This region carries all the necessary elements capable of responding to Hh/Wnt signaling properly. Further analysis on 100-bp-long nested deletions has shown that four deletions decease dlp2.1.5-GFP up-regulation in Hh/Wnt signalingdefective IGS cells, but no single deletion up-regulates dlp2.1.5-GFP expression, suggesting that multiple repressive elements in the regulatory region are required for repressing dlp expression in the niche, likely by antagonizing the function of the activating elements. Consistently, both Pan and Ci can bind to multiple sites of the identified regulatory region in vitro and in vivo. These Pan and Ci binding sites are also overlapped with the regions containing the activating elements. Last, mutating either Pan binding sites or Ci binding sites in the dlp regulatory region causes the moderate up-regulation of dlp2.1.5-GFP in the niche compared with its strong up-regulation in the Hh/Wnt signalingdefective niche. Curiously, these mutations also decrease the expression of dlp2.1.5-GFP in follicle cell progenitors. Together, these findings lead us to propose a model that on Hh and Wnt signaling activation, Ci and Pan bind the regulatory region of dlp and repress its expression in the niche partly by preventing the recruitment of unknown transcriptional activators.

This study has further suggested that Ci and Pan sequentially recruit Croc and Egg/H3K9 to the dlp regulatory region to maintain transcriptional repressive mark H3K9me3 and, thus, prevent dlp transcription. Both Croc and Egg are required in the niche for repressing dlp expression and for promoting GSC progeny differentiation. egg is an H3K9 trimethylase required in the niche for promoting GSC progeny differentiation (9), whereas Croc is a known fork head domaincontaining transcriptional factor (43). Croc can also directly bind to two independent sites in the same dlp regulatory region in an Hh/Wnt signalingdependent manner in vivo, and one of them is also in close proximity to Pan and Ci binding sites. In S2 cells, both Pan and Ci are associated with Croc, which is also associated with and stabilizes Egg. On the basis of these results, we propose that on Hh and Wnt signaling activation, Ci and Pan recruit the Croc-Egg protein complex to the dlp regulatory region to directly repress dlp expression, likely through maintaining H3K9me3 (Fig. 6L).

Among six Dlp-related mammalian GPC proteins, GPC4 and GPC6 can functionally replace Dlp to promote Hh signaling in Drosophila, whereas GPC2, GPC3, and GPC5 are inhibitory on Hh signaling when overexpressed (26). In mammals, Dlp homologs GPC3 and GPC5 can inhibit and activate Hh signaling, respectively (25, 55), whereas GPC3 and GPC4 can promote and repress canonical Wnt signaling (27, 56), indicating that the ability of Dlp to repress and activate Hh and Wnt signaling is conserved from Drosophila to mammals. These findings raise the interesting possibility that the Dlp-mediated feedback control of Hh and Wnt signaling interdependence might also help elucidate their cooperative mechanisms in mammalian development, stem cell regulation, and cancer.

The following Drosophila stocks used in this study are described in FlyBase, unless specified: c587, tubulin-gal80ts, smoRNAi (BL27037 and BL62987) (12), dshRNAi (BL31306 and BL31307), ciRNAi (BL64928), PanRNAi (BL40848), dlpRNAi (BL34089 and BL34091), crocRNAi (BL27071 and BL34647), eggRNAi (BL32445 and BL34803), tkvRNAi (BL40937 and BL57303), lucRNAi (BL31603), bamGFP, Dad-lacZ, ptc-GFP (29, 30), fz3-RFP (28, 30), UAS-CD8::GFP, UAS-dlp, UAS-Croc-3HA (FlyORF: F000139), dlpMI04217, dlpMI09937, and dlpMI10064. Drosophila strains were maintained and crossed at room temperature on standard cornmeal/molasses/agar media unless specified. To maximize the RNAi-mediated knockdown effect, newly eclosed flies at room temperature were cultured at 29C for the specified days before phenotypic analysis.

The Invitrogen Gateway Technology was used to make the constructs for expressing Flag-tagged Ci, Flag-tagged Pan, Flag-tagged Dlp, HA-tagged Wnt2, HA-tagged Wnt4, Flag-tagged Egg, Flag-tagged Croc, and HA-tagged Croc in S2 cells for co-IP experiments or for making transgenic flies. The coding sequences for ci, pan, dlp, wnt2, wnt4, egg, and croc were amplified from Drosophila ovarian cDNAs using PCR. The armS10 sequence was amplified from the genomic DNA from the UAS-armS10 transgenic strain (BL4782). All the PCR products were cloned into the pENTR-TOPO cloning vector and were completely sequenced. These pENTR vectors were subsequently recombined into Flag-, Myc-, or HA-tagged destination vectors (pAWF, pAWH, pAWM, and pTWF) by using LR Clonase (Invitrogen). UAS-Myc-CiPKA was a gift from J. Jiang (57). Since Dlp protein undergoes internal proteolytic cleavage, the 3 Flag tag was inserted after the 18th amino acid residue, and the termination codon was added to the reverse primer to skip the Flag tag in the pAWF destination vector. The GST fusion proteins with Ci (GST-Ci), Pan (GST-Pan), or Croc (GST-Croc) were constructed by cloning the DNA fragments encoding five Ci ZnF_C2H2 domains, the Pan HMG domain, or the Croc FH domain into the Eco RI and Xho I sites of pGEX4T1, respectively.

Ovaries were dissected, fixed, and stained according to the method described previously (58, 59). The following antibodies were used in this study: mouse monoclonal anti-Dlp antibody [1:10; Developmental Studies Hybridoma Bank (DSHB)], mouse monoclonal anti-Hts antibody (1:50; 1B1, DSHB), rabbit polyclonal anti--galactosidase (LacZ) antibody (1:500; MP Biomedical, no. 08559761), mouse monoclonal anti--galactosidase (LacZ) antibody (1:50; JIE7, DSHB), rabbit monoclonal anti-Smad3 antibody (pS423/pS425) (1:200; Epitomics, ab52903), rabbit polyclonal anti-RFP (1:1000; Rockland, no. 600-401-379), and chicken polyclonal anti-GFP antibody (1:500; Invitrogen, no. A10262).

S2 cells were grown at 25C in the HyClone SFX-Insect Cell Culture Media (Thermo Fisher Scientific). Transfections were performed using the X-treme GENE HP (6366546001, Roche) transfection reagent according to the manufacturer's instructions. For co-IP experiments in S2 cells, 12 ml of S2 cells was transfected by indicated plasmids. The transfected S2 cells were then lysed with 800 l of ice-cold lysis buffer [50 mM tris-HCl (pH 7.5), 150 mM NaCl, 0.5% Triton X-100, 1 mM EDTA, and a mixture of protease inhibitors]. The supernatant of the lysate was incubated with 2 g of mouse anti-HA, mouse anti-Flag, or mouse anti-Myc. Protein A/G agarose (40 l; sc-2003; Santa Cruz Biotechnology), which was prewashed in 5% bovine serum albumin at 4C for 1 hour, was added to the supernatant. The supernatant-antibody-agarose mix was incubated overnight at 4C. After six washes with the lysis buffer, the bound complexes were eluted with 2 SDS sample buffer and subjected to SDSpolyacrylamide gel electrophoresis and immunoblotting. Mouse anti-tubulin (T9026, Sigma-Aldrich; 1:10000), mouse anti-Flag (F1804, Sigma-Aldrich; 1:2000), mouse anti-Myc (M4439, Sigma-Aldrich; 1:2000), or mouse anti-HA (H3663, Sigma-Aldrich; 1:2000) antibodies were used for immunoblotting. To avoid the interference of immunoglobulin G (IgG) heavy chain (~55 kDa), horseradish peroxidasegoat anti-mouse IgG light chain secondary antibodies were used. Inputs were extracted before IP.

To construct the dlp reporter plasmids, we then used the following primer pairs carrying either Xba I or Kpn I at the 5 end to amplify the DNA fragments from the Drosophila genomic DNA, which were confirmed by complete sequencing and then cloned into the pGR vector:

dlp promotor: agtctctagactttcgatagtgtggaccttcctt; aagtggtaccgtatgtacagtgtcactaggctat.

dlp2.1: cgactctagagtatgtccgatattatataccaat; cgacggtaccgcatttataactttgttgtagttg.

dlp2.2: cgactctagataataatagtaggca; cgacggtaccttgccacattccaccttagctatt.

dlp2.3: aaggtctagaaatggggctagctta; cgacggtaccaagggagaacggagccaaactcca.

dlp2.4: ttggtctagaacaagttttcgaatga; cgacggtaccatgtggacataatcgagcataa.

dlp2.5: attttctagaatgtatttctggagt; cgacggtaccagactctgatacgcatacaggata.

dlp2.6: agtctctagatggtgccacactcca; aagtggtaccattttgttaatctct.

dlp4: agtctctagagtgagtagtagtctgcgaaatcca; aagtggtacctggaaaataagattaaatcggtg.

dlp6: agtctctagagtgagatctacagcggaataatt; aagtggtacctgcaatgaattaatttgagagtt.

dlp2.1.1: cgactctagagtatgtccgatattatataccaat; cgacggtacctcacgcagttcacgccaacgatgct.

dlp2.1.2: cgactctagaagcatcgttggcgtgaactgcgtga; cgacggtaccaatctgttattaaaatttgtccta.

dlp2.1.3: cgactctagataggacaaattttaataacagatt; cgacggtaccagttgcgatctacaaagccaatct.

dlp2.1.4: cgactctagaagattggctttgtagatcgcaact; cgacggtaccacaatggtcaacaattgcagaagt.

dlp2.1.5: cgactctagaacttctgcaattgttgaccattgt; cgacggtacctggccacgtttgacctgctcgaga.

dlp2.1.6: cgactctagatctcgagcaggtcaaacgtggcca; cgacggtaccgcatttataactttgttgtagttg.

dlp2.1.51: gctctagactgtctggtgtttgtttatgagg; cgacggtaccgcatttataactttgttgtagttg.

dlp2.1.52: gctctagaaaaacttatgaagcttttttaatatgattagcaaac; cgacggtaccgcatttataactttgttgtagttg.

dlp2.1.53: gctctagacatctggtaaaccgaaagctt; cgacggtaccgcatttataactttgttgtagttg.

dlp2.1.54: gctctagatacaattactcagttcctagggg; cgacggtaccgcatttataactttgttgtagttg.

dlp2.1.55: gctctagacggtgctgggattccaga; cgacggtaccgcatttataactttgttgtagttg.

dlp2.1.56: cgactctagaacttctgcaattgttgaccattgt; agacggtacctgccggcaattaagtcgt.

dlp2.1.57: cgactctagaacttctgcaattgttgaccattgt; agacggtacctccacaggattcattcttagaaaatttgc.

dlp2.1.58: cgactctagaacttctgcaattgttgaccattgt; agacggtacctcagctaattacgcgaaattgc.

dlp2.1.59: cgactctagaacttctgcaattgttgaccattgt; agacggtaccatcactggatcagatagcacc.

dlp2.1.510: cgactctagaacttctgcaattgttgaccattgt; agacggtaccatggcatattagggggcg.

To make dlp2.1.5(3Pan*)-GFP transgenes, the three identified Pan binding sites in gcccacaaagtcaacacttgctga, ctgacgatgctgacagaaatggga, and tcagcaaattttctaagaatgaat were mutated to gcccacaaagtttacacttgctga, ctgacgatgcaaacagaaatggga, and tttgcaaattttctaagaatgaat, respectively. To make dlp2.1.5(4 Ci*)-GFP transgenes, the four identified Ci binding sites in cgtttatcacgggggcttttcgca, actgacaacccactaaactagatc, agcaaactctttcacgcgatctcg, and atgggatctcccagccggcagcca were mutated to cgtttatcacggggtttttttgca, actaacaaaacactaaactagatc, agcaaactctttcacgttatcttg, and atggaatctaacagccggcagcca, respectively. For the dlp2.1.5(3Pan*+4Ci*)-GFP reporter, all of the seven binding sites were mutated. All the constructs were inserted into the attp40 site on the second chromosome using PhiC31 integrasemediated transgenesis by Rainbow Transgenic Flies Inc.

The dlp215 (deleting first 800 bp in dlp2.1.5 region) mutant was designed and generated by Rainbow Transgenic Flies Inc. using the CRISPR-Cas9 technology. The following guide RNAs (gRNAs) were used: gRNA1 target, 5-gaattgttgaccattgtatgg; gRNA2 target, 5-ggccaacgacttaattgccgg. Mutants were confirmed by PCR and sequencing. Primers used for PCR identification were 5-gcaaccaccgcatgactatta and 5-gatgggaaagagacagcaact.

The Escherichia coli bacteria strains were transfected with GST, GST-Ci-ZNF, GST-Pan-HMG, or GST-Croc-FH plasmid, and the culture for each bacteria strain was grown to the density of OD650 (optical density at 650 nm) = 0.1 to 0.25. The expression of the fusion proteins was then induced by the addition of 0.2 mM isopropyl--d-thiogalactopyranoside for overnight at 16C. The cells were then harvested and lysed with B-PER with Enzymes Bacterial Protein Extraction Kit (90079, Thermo Fisher Scientific), and the proteins were purified with glutathione agarose (16100, Thermo Fisher Scientific). The in vitro DNA-protein binding assay was performed according to the LightShift Chemiluminescent EMSA Kit (20148, Thermo Fisher Scientific). Glycerol (4.35%), magnesium chloride (5 mM), poly(dI-dC) (50 ng/ml), and NP-40 (0.05%) were included in the binding reaction. For each 20 l of binding assay, 0.1 nM biotin-labeled probe (synthesized by Integrated DNA Technologies) and 10 g of purified GST protein or GST fusion protein were used.

ChIP was performed essentially as described by the Pierce Agarose ChIP Kit (26156, Thermo Fisher Scientific). For each genotype, 200 pairs of ovaries were dissected and then digested with type II collagenase (50D11833; Worthington). The late-stage egg chambers and mature eggs were filtered and removed. Primers used for regular PCR are as follows:

196: acttctgcaattgttgaccattgt; tcctactcgtttatataccccgcc.

91204: gtaggagttgctgtctggtgtttg; ttttagattttatatacccaaagc.

199294: ctaaaacttatgaagcttttttaa; gatctagtttagtgggttgtcagt.

289402: tagatcacctaacatctggtaaac; taatggcatattagggggcgagat.

397512: ccattacaattactcagttcctag; atcccagcaccgatcactggatca.

507602: tgggattccagacattttgcccac; cgtcagctaattacgcgaaattgc.

597710: gcaatttcgcgtaattagctgacg; tcatccgcgatccacaggattcat.

705800: ggatgattcaagttggattcgagt; tgccggcaattaagtcgttggccc.

For comparing the binding affinities of Croc to the dlp regulatory region between WT, smoKD, and dshKD IGS cells, qPCR was performed using PerfeCTa SYBR Green FastMix (Quantabio, no. 022048) according to the manufacturers recommendations and analyzed using the 2CT method. Sequences of primers are tgggattccagacattttgcccac and gtcagctaattacgcgaaattgc. actin5C was used as internal control (primers: atcgggatggtcttgattctg and actccaaacttccaccactc).

To assess the expression level of fz3, ptc, and dlp mRNA in control, smoKD, and dshKD IGS cells, we performed FISH on ovaries, which are immunostained for PZ1444-LacZ (labeling IGS cells). Hybridization chain reaction (HCR) was used to achieve high-sensitivity FISH for quantification. Probe sets against fz3-mRNA (lot: PDR091), ptc-mRNA (lot: PDR092), or dlp-mRNA (lot: PDR093) were ordered from Molecular Instruments Inc. Immunostaining ovaries using anti-LacZ antibodies was performed according to the previous publication before in situ hybridization (60). Then, standard steps following HCR v3.0 protocol for whole-mount fruit fly embryos were applied. At the end of HCR in situ hybridization, 4,6-diamidino-2-phenylindole (DAPI) was added at 1 g/l for 10 min in 5 SSCT (1X saline-sodium citrate buffer with 0.1% Triton X-100) buffer to the ovaries and then washed 15 min in 5 SSCT for four times. Last, the ovaries were mounted, and images were captured according to the regular immunostaining protocol.

GSCs and CBs were quantified under the fluorescence microscope according to the method described previously (58). The germaria were imaged by the Leica SP5 confocal microscope, and the images with all sections were merged unless specified. For confocal images, fluorescence intensities for the highlighted areas of interest were quantified using the Leica software, and the mean values of fluorescence intensities and internal controls were collected. The ratio of mean values of intensities of interest to internal controls was calculated and subjected for statistical analysis using Students t test in Microsoft Excel or GraphPad Prism 7. For fz3-RFP and ptc-GFP reporters, the intensity of single IGS cell nuclear was measured, because these reporters express nuclear located RFP or GFP. For other staining, intensity of IGS cell region in each germarium was measured. All bar graphs are represented as means standard error and with individual value (***P 0.001; **P 0.01; *P 0.05; n.s., no significance).

See more here:
Dlp-mediated Hh and Wnt signaling interdependence is critical in the niche for germline stem cell progeny differentiation - Science Advances