Global Choroideremia Treatment Market Growth, Outlook, Trend and Forecast by Wize Pharma Inc, Spark Therapeutics, Inc, PIXIUM VISION, Retina Implant…

Market Definition:

Choroideremia is also known as choroidal sclerosis is a rare, degenerative, X-linked inherited retinal disorder characterized by progressive degeneration of the choroid, retinal pigment epithelium (RPE) and retina due to Mutations in the CHM gene. This CHM gene required to produce Rab escort protein-1 (REP-1). The condition gets its name from the distinctive sweet odor of affected infants urine and is also c, a protein that takes part in targeting vesicles (small sacs of substances) into, out of, and within cells.

According to the statistics published by U.S. Department of Health & Human Services, an estimated population of choroideremia is about 1 in 50,000-100,000 people. Presence of refined healthcare infrastructure and emerging new market are the key factors for growth of this market.

Global Choroideremia Treatment Market By Treatment Type (Gene Therapy, Surgery), Route of Administration (Oral, Injectable), End- Users (Hospitals, Homecare, Specialty Clinics, Ophthalmic Clinics and Others), Distribution Channel (Hospital Pharmacies, Retail Pharmacies), Geography (North America, South America, Europe, Asia-Pacific, Middle East and Africa) Forecast to 2026

Global Choroideremia Treatment Marketis expected to grow at a steady CAGR in the forecast period of 2019-2026.Increase in strategic alliances between the pharmaceuticals companies and high demand of disease specific novel therapies are the key factors that fueling the market growth.

Some of the major competitors currently working in the global choroideremia treatment market areBiogen, 4D Molecular Therapeutics, Copernicus Therapeutics, Inc, Wize Pharma Inc, Spark Therapeutics, Inc, PIXIUM VISION, Retina Implant AG, F. Hoffmann-La Roche Ltd and others.

Download exclusive PDF sample report @https://www.databridgemarketresearch.com/request-a-sample/?dbmr=global-choroideremia-treatment-market

Market Drivers

Market Restraints

Segmentation:Global Choroideremia Treatment Market

By Treatment

By Route of Administration

By End Users

By Distribution Channel

ByGeography

Download Detailed TOC @https://www.databridgemarketresearch.com/toc/?dbmr=global-choroideremia-treatment-market

Key Developments in the Market:

Competitive Analysis:

Global choroideremia treatment market is highly fragmented and the major players have used various strategies such as new product launches, expansions, agreements, joint ventures, partnerships, acquisitions, and others to increase their footprints in this market. The report includes market shares global choroideremia treatment market for Global, Europe, North America, Asia-Pacific, South America and Middle East & Africa.

Reasons to Purchase this Report

Order a Copy of Global Choroideremia Treatment Market Report @https://www.databridgemarketresearch.com/inquire-before-buying/?dbmr=global-choroideremia-treatment-market

About Data Bridge Market Research

An absolute way to forecast what future holds is to comprehend the trend today!

Data Bridge set forth itself as an unconventional and neoteric Market research and consulting firm with unparalleled level of resilience and integrated approaches. We are determined to unearth the best market opportunities and foster efficient information for your business to thrive in the market. Data Bridge endeavors to provide appropriate solutions to the complex business challenges and initiates an effortless decision-making process. Data Bridge is an aftermath of sheer wisdom and experience which was formulated and framed in the year 2015 in Pune.

Contact:

Data Bridge Market Research

US: +1 888 387 2818

UK: +44 208 089 1725

Hong Kong: +852 8192 7475

Email:Corporatesales@databridgemarketresearch.com

The rest is here:
Global Choroideremia Treatment Market Growth, Outlook, Trend and Forecast by Wize Pharma Inc, Spark Therapeutics, Inc, PIXIUM VISION, Retina Implant...

Juvenile Macular Degeneration (Stargardt Disease) Treatment Market Growth and key Industry Players 2021 Analysis and Forecasts to 2026 – Express…

The Juvenile Macular Degeneration (Stargardt Disease) Treatment market report provides a thorough analysis of this business landscape based on the consumption and production aspects. With respect to consumption, the report reviews the product consumption value as well as the product consumption volume alongside the individual sales trends of each product during the forecast period. In addition, details regarding the import and export graphs across the various geographies are also provided in the report.

According to Latest Research Report on Juvenile Macular Degeneration (Stargardt Disease) Treatment Market size | Industry Segment by Applications (Hospitals,Eye Clinics andOthers), by Type (Stem Cell Therapy,Gene Therapy andOthers), Regional Outlook, Market Demand, Latest Trends, Juvenile Macular Degeneration (Stargardt Disease) Treatment Industry Share, Research Growth Forecast & Revenue by Manufacturers, The Leading Company Profiles, Growth Forecasts 2026.

Request Sample Copy of this Report @ https://www.express-journal.com/request-sample/85233

Based on the production aspect, the report covers the manufacturing of the product, its revenue, and gross margins garnered by the market majors. Variation in unit costs strategized by these manufacturers across various regional markets during the analysis period are also entailed in the report.

A brief of the regional outlook:

An overview of the product spectrum:

A gist of the application terrain:

Insights regarding the competitive terrain:

In summary, the Juvenile Macular Degeneration (Stargardt Disease) Treatment market report is evaluated through several categorizations, including the basic industry definitions. Information pertaining the upstream raw materials, downstream buyers, and distribution channels of the competitors are discussed in the report. The study also examines the key drivers, restraints and opportunities that will impact the growth trends in the ensuing years.

Highlights of the Report:

Key Questions Answered in the Report Include:

Request Customization on This Report @ https://www.express-journal.com/request-for-customization/85233

Link:
Juvenile Macular Degeneration (Stargardt Disease) Treatment Market Growth and key Industry Players 2021 Analysis and Forecasts to 2026 - Express...

Size & Share Of Gene Therapy Market 2020 Report Including COVID-19 Impact Analysis And Forecast Till 2026 – Northwest Trail

Facts & Factors Market Research added a recent report on Connected Medical Devices Security Market By Technology (Wired, Wireless, and Hybrid), By Security Type (Cloud Security, Network Security, Wireless Security, and Application Security), By Category (Telemetry Systems, Integration/Networking Platforms, and Interface Devices), and By End-User (Clinics, Laboratories, Hospitals, Diagnostic Centers, and Home Healthcare): Global Industry Perspective, Comprehensive Analysis, and Forecast, 2018 2025 to its research database. The Gene Therapy Market research report is an output of a brief assessment and an extensive analysis of practical data collected from the global industry.

This specialized and expertise oriented industry research report scrutinizes the technical and commercial business outlook of the Gene Therapy industry. The report analyzes and declares the historical and current trends analysis of the Gene Therapy industry and subsequently recommends the projected trends anticipated to be observed in the Gene Therapy market during the upcoming years.

The Gene Therapy market report analyzes and notifies the industry statistics at the global as well as regional and country levels to acquire a thorough perspective of the entire Gene Therapyt market. The historical and past insights are provided for FY 2016 to FY 2019 whereas projected trends are delivered for FY 2020 to FY 2026. The quantitative and numerical data is represented in terms of value from FY 2016 2026.

The quantitative data is further underlined and reinforced by comprehensive qualitative data which comprises various across-the-board market dynamics. The rationales which directly or indirectly impact the Gene Therapy industry are exemplified through parameters such as growth drivers, restraints, challenges, and opportunities among other impacting factors.

Request an Exclusive Free Sample Report of Gene Therapy Market: http://www.fnfresearch.com/sample/gene-therapy-market-by-type-germ-line-gene

Our Every Free Sample Includes:

A research report overview, TOC, list of tables and figures, an overview of major market players and key regions included.

Some of Top Market Players Analysis Included in this Report:

The Market Player Analysis based on some of below Factors:

This research report provides forecasts in terms of CAGR, and Y-O-Y growth. This helps to understand the overall market and to recognize the growth opportunities in the global Gene Therapy Market. The report also includes a detailed profile and information of all the major market players currently active in the global Gene Therapy Market. The companies covered in the report can be evaluated based on their latest developments, financial and business overview, product portfolio, key trends in the market, long-term and short-term business strategies by the companies to stay competitive in the market.

The global Gene Therapy Market size & trends are classified based on the types of products, application segments, and end-user. Each segment expansion is assessed together with the estimation of their growth in the upcoming period. The related data and statistics collected from the regulatory organizations are portrayed in the Gene Therapy Market report to assess the growth of each segment.

The global Gene Therapy Market size & trends are classified based on the types of products, application segments, and end-user. Each segment expansion is assessed together with the estimation of their growth in the upcoming period. The related data and statistics collected from the regulatory organizations are portrayed in the Gene Therapy Market report to assess the growth of each segment.

Request Customized Copy of Report @ http://www.fnfresearch.com/customization/gene-therapy-market-by-type-germ-line-gene

(We customize your report according to your research need. Ask our sales team for report customization).

Available Customization Options:

The Gene Therapy Market can be customized to the country level or any other market segment. Besides this, Report understands that you may have your own business need, hence we also provide fully customized solutions to clients.

KEY REPORT POINTERS & HIGHLIGHTS:

To Know More about Discount on Specific Report, Kindly Mail us at [sales@fnfresearch.com] with Your requirement. We will send you a quote and discount offer, if available.

About Us:

Facts & Factors is a leading market research company and offers customized research reports and consulting services. Facts & Factors aims at management consulting, industry chain research, and advanced research to assist our clients by providing planned revenue model for their business. Our report and services are used by prestigious academic institutions, start-ups, and companies globally to understand the international and regional business background. Our wide-ranging database offers statistics and detailed analysis of different industries worldwide that help the clients in achieving sustainable progress. The well-organized reports help clients in developing strategies and making informed business decisions.

Contact Us:

Facts & Factors

Global Headquarters

Level 8, International Finance Center, Tower 2,

8 Century Avenue, Shanghai,

Postal 200120, China

Tel: +8621 80360450

E-Mail: sales@fnfresearch.com

Web: http://www.fnfresearch.com

View post:
Size & Share Of Gene Therapy Market 2020 Report Including COVID-19 Impact Analysis And Forecast Till 2026 - Northwest Trail

Fulcrum Therapeutics, Inc. (FULC) Q1 2020 Earnings Call Transcript – Motley Fool

Image source: The Motley Fool.

Fulcrum Therapeutics, Inc.(NASDAQ:FULC)Q12020 Earnings CallMay 13, 2020, 8:00 a.m. ET

Operator

Good morning, and welcome to Fulcrum Therapeutics first-quarter 2020 conference call. [Operator instructions] I would now like to turn the call over to Christi Waarich, director of investor relations and corporate communications at Fulcrum. Please proceed.

Christi Waarich -- Director of Investor Relations and Corporate Communications

Thank you, Dmitria. Good morning, and welcome to the Fulcrum Therapeutics conference call to discuss our first-quarter 2020 financial results and recent corporate highlights. Earlier today, we issued a press release outlining our recent progress. For those of you who don't have a copy, you can access it in the investor relations section of our website fulcrumtx.com.

Please be reminded that remarks made during this call may contain forward-looking statements within the meaning of the Private Securities Litigation Reform Act of 1995. These may include statements about our future expectations and plans, clinical development time lines and financial projections. While these forward-looking statements represent our views as of today, they should not be relied upon as representing our views in the future. We may update these statements in the future but we are not taking on an obligation to do so.

Please refer to our most recent filings with the Securities and Exchange Commission for a discussion of certain risks and uncertainties associated with our business. With me on today's call are Robert Gould, president and chief executive officer; Diego Cadavid, senior vice president of clinical development; Owen Wallace, chief scientific officer; and Bryan Stuart, chief operating officer. Let me quickly run through this morning's agenda. Robert will begin the call with an overview of our recent progress.

Diego will discuss our FSHD program. Owen will discuss our sickle cell program and research efforts, and Bryan will cover our financials before opening the call for Q&A. With that, it's my pleasure to turn the call over to Robert. Robert?

Robert Gould -- President and Chief Executive Officer

Thank you, Christi. Good morning, everyone, and thank you for joining us today. I first want to thank the healthcare workers, investigators and caregivers for their courage and passion as they continue to support so many during the challenges of COVID-19. Our hearts go out to everyone who's been impacted.

To all of our friends, colleagues and the patient communities we serve, we hope you are keeping safe and healthy. Fulcrum's mission and purpose remain unchanged as we work to discover and develop therapeutics to treat genetically defined diseases by addressing their root cause. I'm proud of how our employees have risen to the evolving challenges of the COVID-19 pandemic. I would like to begin by highlighting some of our recent updates and accomplishments.

Today, we announced an amendment to ReDUX4, our Phase 2b trial with losmapimod in patients with facioscapulohumeral muscular dystrophy or FSHD. Diego will go over the amendment in more detail. These changes will extend the patient treatment from the original trial design and we believe will provide a more robust data set while addressing the challenges presented by COVID-19. Early in the quarter, we received orphan drug designation from the U.S.

Food and Drug Administration for losmapimod in FSHD. I'm pleased to report that we have also received orphan designation from the European Commission for losmapimod for the treatment of FSHD. Like in the U.S., orphan designation is granted by the European Commission to drugs that are intended for the treatment, prevention or diagnosis of life-threatening or chronically debilitating rare diseases. We are extremely pleased to have received this designation, further supporting the advancement of losmapimod's FSHD program.

We recently presented dose-dependent target engagement data in skeletal muscle from our Phase 1 trial with losmapimod during a virtual clinical trial session of the muscular dystrophy association meeting. We continue to make progress with FTX-6058, an oral small-molecule therapeutic designed to induce expression of fetal hemoglobin in select hemoglobinopathies. You'll hear about our sickle cell program from Owen later in the call. We also continue to make progress on our early research-stage activities, including building out FulcrumSeek, our proprietary product engine designed to identify drug targets, programs and clinical development candidates in a broad range of genetically defined diseases.

And we initiated research activities under our collaboration with Acceleron. I would now like to turn the call over to Diego for an update on the FSHD program. Diego?

Diego Cadavid -- Senior Vice President of Clinical Development

Thanks, Robert. As a reminder, FSHD is a progressive disease characterized by severe muscular degeneration that occurs as skeletal muscle is replaced by fat. We estimate there are approximately 16,000 to 38,000 patients in the U.S. and similar incidents worldwide.

There are currently no approved drugs for FSHD and we are advancing the only known industry-sponsored clinical trial evaluating a potential treatment. Unlike other diseases that can be characterized by the lack of a gene, FSHD is characterized by the aberrant expression of the gene DUX4, the root cause of the disease. We at Fulcrum discovered that losmapimod, a selective p38 MAP kinase inhibitor, reduced the expression of DUX4 in preclinical studies. We therefore believe losmapimod represents a potential novel therapeutic option for FSHD patients.

Our own evidence, as well as independent evidence suggests that we do not have to turn DUX4 off completely to provide benefit. There is a spectrum of DUX4 expression and FSHD presentation that suggests that even an incremental reduction may be beneficial to patients. Thus, we believe, as do independent researchers, that any reduction in DUX4-driven gene expression has the potential for benefit to patients. ReDUX4 is our international Phase 2b, double-blind placebo-controlled trial of losmapimod in patients with genetically confirmed FSHD.

We completed enrollment of 80 patients at the end of February, which exceeded the 66 we had originally planned. The primary endpoint of the trial is the change from baseline in DUX4-driven gene expression in affected skeletal muscle. We also completed enrollment in our Phase 2 single-site open-label trial which has been impacted by COVID-19, and we are considering next steps. Fulcrum is dedicated to maintaining the highest standards in patient and clinician safety in the planning and execution of our clinical research programs.

The safety of our clinical trial participants and their healthcare providers, as well as the integrity of the data we collect remains paramount. In the wake of COVID-19, a number of our clinical trial sites postponed trial-related activities, and we quickly implemented plans to limit the potential disruption to our FSHD program. The original design of the ReDUX4 included a pretreatment biopsy followed by a second biopsy at week 16 of the 24-week treatment period. Following the 24-week trial, patients had the opportunity to roll into an open-label extension.

Prior to the COVID-19 pandemic, 12 of the 80 patients completed their 24 weeks of treatment, including their week 16 biopsy and all enrolled in the open-label extension. As the COVID-19 pandemic continues, our team, in collaboration with our investigators, extended the ReDUX4 trial from 24 to 48 weeks. This allows approximately 67 subjects currently continuing in the trial to receive a biopsy at either week 16 or under the amended protocol at week 36 and after completing the 48-week treatment period, rolling to the open-label extension. To summarize, the ReDUX4 trial has been extended from 24 to 48 weeks with an open-label extension to follow.

Patients will receive a muscle biopsy at either 16 or 36 weeks. This extension will apply to the approximately 67 patients still enrolled in the trial while 12 have already completed and have been rolled into the open-label extension. As part of the modification to the trial, we will also conduct an interim analysis of approximately 25 subjects who have completed their 16-week biopsy. We anticipate sharing data from this interim analysis of subjects' DUX4-driven gene expression signature in the third quarter of this year, and we expect to report top-line data on the primary endpoint in the first quarter of 2021.

The extension from 24 to 48 weeks also allows for a longer assessment in a placebo-controlled design of the skeletal muscle MRI secondary endpoint and the various exploratory clinical endpoints such as reachable workspace, FSHD Timed Up and Go, muscle function measures and patient-reported outcomes. From both independent researchers, as well as our own preparatory studies, we know the DUX4 gene signature is stable over time in this population, and we believe that the longer we are able to treat patients, the greater the potential benefit losmapimod may have on the root cause of the disease. We strongly believe these changes to the ReDUX4 study are in the best interest of the patient community and provide the best opportunity to advance this important development effort as we work to address the challenges presented by COVID-19. All of these changes are designed to enable patients and investigators to continue participation in ReDUX4 and will allow us to collect essential data to support continued dialogue with regulators.

I'll now turn the call over to Owen. Owen?

Owen Wallace -- Chief Scientific Officer

Thanks, Diego. At Fulcrum, we pursue targeted indications where we believe we can develop safe and effective small-molecule therapies to rebalance gene expression. In our work across various indications, we consistently aim to address the root causes of disease to increase the potential efficacy of these treatments and, more broadly, transform the way these diseases are being treated. In spite of the challenges posed by COVID-19, we have continued to make progress on our research and early clinical portfolio.

As an essential business, we continue lab operations, albeit on a more limited basis. As a result, we continue to advance the collaboration with Acceleron, as well as our internal portfolio. We have also advanced our work on FulcrumSeek, our proprietary product engine designed to identify drug targets, programs and clinical development candidates in a broad range of genetically defined diseases. By combining high-throughput RNA sequencing, cellular imaging data and large-scale machine learning, we are monitoring more than 10,000 molecular and cellular features generated by the small-molecule probe and CRISPR perturbagen libraries.

Understanding their effects on gene expression is fundamental to our therapeutic strategy to modulate the genetic root cause of disease. FulcrumSeek is not only the core of our target identification strategy. It also provides us with a unique understanding of how cellular function is altered in human disease. I would like to thank our employees who have continued to work diligently through the COVID-19 crisis to advance our research programs, especially those who are coming into the lab working under social distancing and enhanced health and safety guidelines.

Likewise, our hemoglobinopathy program has continued to advance toward the IND filing. Our approach has focused on the up-regulation of fetal hemoglobin, which could be beneficial for both sickle cell disease and beta-thalassemia. By increasing levels of HbF to compensate for the mutated hemoglobin in sickle cell patients, we believe that we can develop and deliver a potent, effective and selective therapy for patients. This therapeutic strategy is supported by human genetics and pharmacology data where increasing levels of HbF have been shown to be associated with improved prognosis and outcomes, suggesting that HbF may be a surrogate endpoint in future clinical trials.

We're very pleased with our recent progress. Our clinical candidate FCX-6058 has been profiled broadly in preclinical in-vitro and in-vivo models of sickle cell disease, and we have seen robust elevation of HbF at drug concentrations that we believe will be readily achieved in humans based on pharmacokinetic profiling of the compound. We've had an abstract accepted for oral presentation at the 14th Annual Sickle Cell Disease Research & Educational Symposium scheduled for September of this year. We have also filed our non-provisional patent application, and we've completed our IND-enabling studies and toxicology work with FTX-6058.

We plan to submit the IND in sickle cell disease in the second half of 2020 and initiate our Phase 1 trial by the end of the year.With that, I will now turn the call over to Bryan for an update on our financial results for the quarter. Bryan?

Bryan Stuart -- Chief Operating Officer

Thanks, Owen. In these unprecedented times, Fulcrum is committed to making a difference for patients with FSHD and select hemoglobinopathies such as sickle cell disease. We are proceeding with a great sense of urgency to bring these potentially transformative therapies to patients. We ended the first-quarter 2020 with $81.2 million in cash, cash equivalents and marketable securities.

Based on our current operating plan and projections, we believe this will support our operations into the third quarter of 2021, allowing us to advance losmapimod in FSHD and bring FTX-6058 into the clinic while continuing to invest in our discovery-stage efforts. Research and development expenses for the quarter ended March 31, 2020, were $14.5 million, compared to $34.6 million in the first quarter of 2019. Included in that $34.6 million was $25.6 million of onetime costs associated with the issuance of series B convertible preferred stock under the company's license agreement with GSK for the rights to losmapimod. Excluding these onetime costs, the increase of $5.5 million was primarily due to increased costs related to the advancement of losmapimod for the treatment of FSHD, as well as increased personnel-related costs to support the growth of Fulcrum's research and development organization.

General and administrative expenses for the first quarter of 2020 were $5.1 million as compared to $2.6 million for the first quarter of 2019. This increase was primarily due to increased personnel-related costs to support the growth of our organization, as well as increased costs associated with operating as a public company. Overall, we remain undeterred in our mission and continue to expect several upcoming catalysts. We'll report the interim analysis from ReDUX4 in the third quarter of this year.

We'll initiate the Phase 1 trial with FTX-6058 in sickle cell disease and disclose the biochemical drug target by the end of the year, and we'll continue to advance our discovery programs from our product engine while making progress with our partners at Acceleron. We're excited about the work ahead and we continue -- as we continue to execute on our plans, and we look forward to keeping you updated on our progress in the months ahead. Operator, you may now open the line for questions.

Christi Waarich -- Director of Investor Relations and Corporate Communications

Operator, we're now ready for questions.

Operator

[Operator instructions] And our first question comes from Matthew Harrison with Morgan Stanley. You may proceed.

Kostas Biliouris -- Morgan Stanley -- Analyst

This is Kostas on for Matthew. A couple of questions from my side. The first one is whether you guys expect to lose any power given that you will only have 25 subjects in the first interim analysis. Do you think you will have enough power to see a signal there? Or do you expect the data only to be directional, to show you an improvement or not?

Diego Cadavid -- Senior Vice President of Clinical Development

Yeah. Thank you for the question. This is Diego Cadavid. The sample size of 80 subjects is -- we believe has appropriate power to answer the question at the end of the trial.

25 subjects, we believe, will give us an initial opportunity to look at the data and help us prepare and make some early insights into Phase 3 planning.

Kostas Biliouris -- Morgan Stanley -- Analyst

OK, thank you. And a follow-up question. Will you need to recruit additional subjects or you believe you have all the subjects you need at this point?

Diego Cadavid -- Senior Vice President of Clinical Development

We have completed recruitment. We believe we have all the subjects we needed.

Kostas Biliouris -- Morgan Stanley -- Analyst

OK. And then finally, I was wondering whether -- in the second part, when you expect all the subjects to have a biopsy at 16 or 36 weeks, given that there might be a second wave of the pandemic, of additional -- a second wave of infections, how certain you are you can have all the subjects complete the second biopsy at 36 weeks and whether there is any actions you are taking to mitigate this risk of losing some patients there again? Thank you.

Diego Cadavid -- Senior Vice President of Clinical Development

Yeah. When we amended the protocol, we carefully considered exactly what you're referring to. So we've built some windows -- time windows around the 36th week and sites have flexibility, as well as patients. So right now, we anticipate that we will get the data either at week 16 or at week 36 regardless.

Kostas Biliouris -- Morgan Stanley -- Analyst

OK. Thank you very much.

Operator

And our next question comes from Joseph Schwartz with SVB Leerink. You may proceed.

Joseph Schwartz -- SVB Leerink -- Analyst

My question would be, can you talk about how you arrived at a doubling in duration for the ReDUX protocol with respect to the clinical endpoints? Will patients in ReDUX still be evaluated at 24 weeks? And how many patients are hitting this time point in the second half of this year when it seems like social distancing might relax? And then when would most patients be hitting the 48-week time point? Have you done an analysis there to consider that this is in your best interest given -- however this pandemic might evolve with respect to its different waves based on where you're enrolling these patients?

Diego Cadavid -- Senior Vice President of Clinical Development

Yeah. The ReDUX4 trial completed enrollment in about six months between August of last year and February of this year. Therefore, the patients are moving across all the visits over a period of six months. We decided to extend the study by an additional 24 weeks because we believe, based on what is happening and what we expect to happen with COVID-19, this will give flexibility for the patients to collect data across a much longer period, where we expect the clinics to be open even if intermittently.

So overall, we believe that even if some 24-week visits are missed, patients would come back later. And as you know, FSHD is a slowly progressive disease. We are not counting acute events. So as long as we are collecting the data over time, we believe we'll be able to answer the efficacy questions.

Especially, many sites are still open. The impact of the pandemic is not affecting every site.

Joseph Schwartz -- SVB Leerink -- Analyst

And are you able to bring patients in and just strike while the iron is a little bit warmer in this period we seem to be entering as we speak now? Could you bring patients in for an evaluation? Can you talk about -- is it just at 24 and 48 weeks that the clinical assessments are being performed? Or do you have any ability to sneak in some additional sites without making additional protocol adjustments that might require you to take alpha hits?

Diego Cadavid -- Senior Vice President of Clinical Development

Yeah. This amendment builds flexibility. So all the visits of the original protocol over 24 weeks are open -- sites that are open, patients are coming. And the amendment provides additional opportunities at week 36, week 48 with extended windows.

So we really give opportunities to capture as much data regardless of what happens with COVID-19. We're very fortunate that not only the sites but the patients are very committed, and that's reflected in the high subject retention we have on the trial.

Joseph Schwartz -- SVB Leerink -- Analyst

That's very helpful. And then have you been able to garner any insights to date from the open-label trial? It sounds like you suggested it's been impacted from COVID-19, and I heard you're evaluating the next steps there. So why has that been impacted more, it sounds, than ReDUX4? Can you provide any color on that front?

Diego Cadavid -- Senior Vice President of Clinical Development

Yes. The open-label study is single site so you don't have this opportunity we have in ReDUX4 where we have many sites. And therefore, if one region that happens to be where this site is, is heavily affected, of course, the impact will be larger. That site is in the Netherlands.

We have always considered that a learning trial. The trial began in August. So obviously, we've had valuable learnings from that trial, which has always been the goal to inform what we do in ReDUX4. So in that sense, we believe this trial has been helpful.

Joseph Schwartz -- SVB Leerink -- Analyst

That's helpful. Thanks for the color.

Operator

And our next question comes from Tazeen Ahmad with Bank of America. You may proceed.

Tazeen Ahmad -- Bank of America Merrill Lynch -- Analyst

I just wanted to clarify your powering assumptions. So you previously said that the study would be powered to show a 50% reduction of DUX4 at week 16. Just based on the changes that you're talking about, how does that affect the potential path to accelerated approval? And have you spoken with FDA about this particular item?

Robert Gould -- President and Chief Executive Officer

Hi, Tazeen. This is Robert. Just a slight correction on -- I don't believe that we did power the study for a 50% reduction in the DUX4. That was not one of the original assumptions.

But I'll let Diego speak to the powering assumptions we made.

Diego Cadavid -- Senior Vice President of Clinical Development

Yeah. Robert is correct. We have never disclosed what the assumptions are for the power. This amendment is not impacting the power.

The sample size is the same. It only adds some flexibility. Because they're on treatment, muscle biopsy can be at week 16 or week 36, and we don't really expect a loss of subjects based on this amendment. Therefore, nothing has changed about the power assumptions.

Tazeen Ahmad -- Bank of America Merrill Lynch -- Analyst

OK. And how are you taking into account -- you're effectively increasing the length of the study to a year. What are you seeing in compliance rates for the study so far? And does this increase -- do you have any buffer, if you will, for potential dropouts in the study with the extended time of observation?

Robert Gould -- President and Chief Executive Officer

Yeah. Thanks, Tazeen. This is Robert again. One of the things that we've really been struck with is the cooperation of the patients and their willingness to take losmapimod.

Just to remind you, as you know, it's an oral drug taken twice daily, 7.5 milligram tablets, so two tablets in the morning, two tablets in the evening. And we just had not only a high retention rate of the patients, but we believe high compliance as well. And so the original trial was enrolling 66 patients. And because of the response we had from the patient community and the opportunity we had, we actually increased that to 80 patients.

So even if things were to change with the patients, we do believe that we're still going to be able to have the original 66 patients. But at this point in time, we believe we're going to be able to retain most of the patients that are currently in the study, if not all of them that are currently in the study.

Tazeen Ahmad -- Bank of America Merrill Lynch -- Analyst

OK. And my last question is about taking the biopsy at 16 weeks or 36. How did you come up with 36? And how do you feel confident in the integrity of the readings of both time periods? Because there's a big gap between the two.

Diego Cadavid -- Senior Vice President of Clinical Development

Yeah. This is Diego. So we have done our own preparatory study to look at the stability and variability of the DUX4 gene signature and the natural history, and that was done about six, eight weeks apart. The academic group of the Wellstone collaboration had done it over a year apart, and they were very generous and shared all their data with us.

So we know from these two studies that these DUX4 signature at the population level is very stable. So this interval between eight weeks or a year apart basically gives us a good argument that as long as you -- we collect repeated biopsies within that interval, we don't expect any impact on greater variability or loss of signature. So 36 really came in terms of building flexibility for patients and sites who had not obtained a 16-week biopsy as the pandemic moves, assuming that over time there will be a decrease of peaks and sites will be able to reopen and bring the patients in to obtain these biopsies. It's 36 weeks but we have a window so sites and patients can be flexible, and we believe that is the best chance to collecting the efficacy endpoint without losing power and keeping the quality.

Operator

[Operator instructions] And our next question comes from Ted Tenthoff with Piper Sandler. You may proceed.

Continue reading here:
Fulcrum Therapeutics, Inc. (FULC) Q1 2020 Earnings Call Transcript - Motley Fool

Global Stem Cell Therapy Market Forecast & Opportunities, 2025 – ResearchAndMarkets.com – Business Wire

DUBLIN--(BUSINESS WIRE)--The "Global Stem Cell Therapy Market By Type (Allogeneic, Autologous, Syngeneic), By Source of Stem Cells (Adipose Tissue, Bone Marrow, Neural, Embryo/Cord Blood derived, iPSCs, Others), By Application, By End Users, By Region, Forecast & Opportunities, 2025" report has been added to ResearchAndMarkets.com's offering.

The Global Stem Cell Therapy Market is expected to grow at a formidable rate of around 12% during the forecast period. The industry is segmented based on type, source of stem cells, application, end-users, company and region.

The market is driven by the growing popularity and awareness pertaining to the use of stem cells for the prevention and cure of certain life threatening diseases. Additionally, increase in number of stem cell banks and growing investments by the government and private organizations for the development of stem cell preservation infrastructure is further propelling the market across the globe.

Based on type, the market can be categorized into allogeneic, autologous and syngeneic. The allogenic type segment is expected to register the highest growth during forecast period attributable to the rising commercialization of allogeneic stem cell therapy products, wider therapeutic applications of allogeneic stem cells, easy production scale-up process, growing number of clinical trials related to allogeneic stem cell therapies, among others.

Based on end-users, the market can be bifurcated into hospitals and clinics. The hospitals segment is expected to dominate the market during the forecast years. This can be accredited to the rising preference for stem cell therapies offered by hospitals proves beneficial for the business growth. Hospitals have affiliations with research laboratories and academic institutes that carry out research activities for developing stem cell therapies. On introduction and approval of any novel stem therapy, hospitals implement it immediately.

Regionally, the stem cell therapy market has been segmented into various regions namely Asia-Pacific, North America, South America, Europe, and Middle East & Africa. Among these regions, North America is expected to dominate the overall stem cell therapy market during the next five years on account of the increasing number of clinical trials for stem cell-based products and increasing public-private funding & research grants.

Major players operating in the Global Stem Cell Therapy Market include Osiris Therapeutics, Inc., MEDIPOST Co., Ltd., Anterogen Co., Ltd., Pharmicell Co., Ltd., Holostem Terapie Avanzate S.r.l., JCR Pharmaceuticals Co., Ltd., NuVasive, Inc., RTI Surgical, Inc., AlloSource, Thermo Fisher Scientific and others. The companies are developing advanced technologies and launching new services in order to stay competitive in the market.

Years considered for this report:

Objective of the Study

Key Topics Covered

1. Product Overview

2. Research Methodology

3. Executive Summary

4. Voice of Customer

5. Global Stem Cell Therapy Market Outlook

5.1. Market Size & Forecast

5.1.1. By Value

5.2. Market Share & Forecast

5.2.1. By Type (Allogeneic, Autologous, Syngeneic)

5.2.2. By Source of Stem Cells (Adipose Tissue, Bone Marrow, Neural, Embryo/Cord Blood Derived, iPSCs, Others)

5.2.3. By Application (Musculoskeletal, Wound & Injury, Cardiovascular Disease (CVD), Surgery, Acute Graft-Versus-Host Disease, Drug Discovery & Development, Others)

5.2.4. By End Users (Hospitals v/s Clinics)

5.2.5. By Company (2019)

5.2.6. By Region

5.3. Product Market Map

6. Asia-Pacific Stem Cell Therapy Market Outlook

7. Europe Stem Cell Therapy Market Outlook

8. North America Stem Cell Therapy Market Outlook

9. South America Stem Cell Therapy Market Outlook

10. Middle East and Africa Stem Cell Therapy Market Outlook

11. Market Dynamics

11.1. Drivers

11.2. Challenges

12. Market Trends & Developments

13. Competitive Landscape

13.1. Osiris Therapeutics, Inc.

13.2. MEDIPOST Co. Ltd.

13.3. Anterogen Co. Ltd.

13.4. Pharmicell Co. Ltd.

13.5. Holostem Terapie Avanzate S.r.l.

13.6. JCR Pharmaceuticals Co. Ltd.

13.7. NuVasive, Inc.

13.8. RTI Surgical, Inc.

13.9. AlloSource

13.10. Thermo Fisher Scientific

14. Strategic Recommendations

For more information about this report visit https://www.researchandmarkets.com/r/hmawq6

See the original post here:
Global Stem Cell Therapy Market Forecast & Opportunities, 2025 - ResearchAndMarkets.com - Business Wire

Induced Pluripotent Stem Cells Market Growth by Top Companies, Trends by Types and Application, Forecast to 2026 – Cole of Duty

Reprocell

Moreover, the Induced Pluripotent Stem Cells report offers a detailed analysis of the competitive landscape in terms of regions and the major service providers are also highlighted along with attributes of the market overview, business strategies, financials, developments pertaining as well as the product portfolio of the Induced Pluripotent Stem Cells market. Likewise, this report comprises significant data about market segmentation on the basis of type, application, and regional landscape. The Induced Pluripotent Stem Cells market report also provides a brief analysis of the market opportunities and challenges faced by the leading service provides. This report is specially designed to know accurate market insights and market status.

By Regions:

* North America (The US, Canada, and Mexico)

* Europe (Germany, France, the UK, and Rest of the World)

* Asia Pacific (China, Japan, India, and Rest of Asia Pacific)

* Latin America (Brazil and Rest of Latin America.)

* Middle East & Africa (Saudi Arabia, the UAE, , South Africa, and Rest of Middle East & Africa)

To get Incredible Discounts on this Premium Report, Click Here @ https://www.marketresearchintellect.com/ask-for-discount/?rid=219183&utm_source=NYH&utm_medium=888

Table of Content

1 Introduction of Induced Pluripotent Stem Cells Market

1.1 Overview of the Market1.2 Scope of Report1.3 Assumptions

2 Executive Summary

3 Research Methodology

3.1 Data Mining3.2 Validation3.3 Primary Interviews3.4 List of Data Sources

4 Induced Pluripotent Stem Cells Market Outlook

4.1 Overview4.2 Market Dynamics4.2.1 Drivers4.2.2 Restraints4.2.3 Opportunities4.3 Porters Five Force Model4.4 Value Chain Analysis

5 Induced Pluripotent Stem Cells Market, By Deployment Model

5.1 Overview

6 Induced Pluripotent Stem Cells Market, By Solution

6.1 Overview

7 Induced Pluripotent Stem Cells Market, By Vertical

7.1 Overview

8 Induced Pluripotent Stem Cells Market, By Geography

8.1 Overview8.2 North America8.2.1 U.S.8.2.2 Canada8.2.3 Mexico8.3 Europe8.3.1 Germany8.3.2 U.K.8.3.3 France8.3.4 Rest of Europe8.4 Asia Pacific8.4.1 China8.4.2 Japan8.4.3 India8.4.4 Rest of Asia Pacific8.5 Rest of the World8.5.1 Latin America8.5.2 Middle East

9 Induced Pluripotent Stem Cells Market Competitive Landscape

9.1 Overview9.2 Company Market Ranking9.3 Key Development Strategies

10 Company Profiles

10.1.1 Overview10.1.2 Financial Performance10.1.3 Product Outlook10.1.4 Key Developments

11 Appendix

11.1 Related Research

Get Complete Report

@ https://www.marketresearchintellect.com/need-customization/?rid=219183&utm_source=NYH&utm_medium=888

About Us:

Market Research Intellect provides syndicated and customized research reports to clients from various industries and organizations with the aim of delivering functional expertise. We provide reports for all industries including Energy, Technology, Manufacturing and Construction, Chemicals and Materials, Food and Beverage and more. These reports deliver an in-depth study of the market with industry analysis, market value for regions and countries and trends that are pertinent to the industry.

Contact Us:

Mr. Steven Fernandes

Market Research Intellect

New Jersey ( USA )

Tel: +1-650-781-4080

Tags: Induced Pluripotent Stem Cells Market Size, Induced Pluripotent Stem Cells Market Trends, Induced Pluripotent Stem Cells Market Growth, Induced Pluripotent Stem Cells Market Forecast, Induced Pluripotent Stem Cells Market Analysis Sarkari result, Government Jobs, Sarkari naukri, NMK, Majhi Naukri,

Our Trending Reports

Allulose Market Size, Growth Analysis, Opportunities, Business Outlook and Forecast to 2026

Breast Lesion Localization Methods Market Size, Growth Analysis, Opportunities, Business Outlook and Forecast to 2026

See the rest here:
Induced Pluripotent Stem Cells Market Growth by Top Companies, Trends by Types and Application, Forecast to 2026 - Cole of Duty

Citius Announces Data on NoveCite Mesenchymal Stem Cells (NC-MSCs) to be Presented at the American Society of Gene and Cell Therapy (ASGCT) Annual…

- Induced pluripotent stem cell (iPSC)-derived MSCs demonstrate higher-level secretion of anti-inflammatory proteins and greater expansion potential than conventional, donor-derived MSCs

- Data include comparative therapeutic benefit in an experimental autoimmune encephalomyelitis (EAE) mouse model

CRANFORD, N.J., May 13, 2020 /PRNewswire/ -- Citius Pharmaceuticals, Inc. ("Citius" or the "Company") (Nasdaq: CTXR), a specialty pharmaceutical company focused on developing and commercializing critical care drug products, today announced that data on NoveCite MSCs will be presented this week at the American Society of Gene and Cell therapy (ASGCT) annual meeting. NC-MSCs are made by Novellus, Inc. ("Novellus"), a Cambridge-based biotechnology company, using its patented mRNA-based cell-reprogramming process. Earlier this year, Citius signed an exclusive option agreement to in-license NC-MSCs for acute respiratory distress syndrome (ARDS), including in COVID-19 patients, from Novellus.

The data to be presented show that NC-MSCs secrete higher levels of anti-inflammatory proteins compared to MSCs derived from bone marrow. From the abstract: "Comparative secretome analysis showed overexpression of multiple neuroprotective and anti-inflammatory factors, including CXCL1, VEGF-A, and CXCL5." In addition, NC-MSCs showed therapeutic benefit in an experimental autoimmune encephalomyelitis (EAE) mouse model, delaying disease progression and improving the clinical score compared to the control group, while bone marrow-derived MSCs showed no difference from the control.

"We are pleased to present these data at the annual meeting of the ASGCT," said Matt Angel, PhD, co-founder and Chief Science Officer at Novellus. "While conventional MSCs have shown promise in the treatment of inflammatory lung disease, protein secretion and manufacturability remain challenges for these approaches. The data that will be presented this week show that iPSC-derived MSCs secrete higher levels of anti-inflammatory proteins, and exhibit greater expansion potential than bone marrow-derived MSCs. We believe that these properties make iPSC-derived MSCs especially well-suited for an allogeneic cell therapy for ARDS."

"Last month Citius signed an exclusive option agreement with Novellus for worldwide development and commercial rights related to the use of these uniquely derived MSCs for the treatment of ARDS. The pre-clinical data that is being presented at the ASGCT annual meeting adds to our confidence in the higher potency of these MSCs, which we expect will result in better outcomes for patients with COVID-19 and ARDS," stated Myron Holubiak, CEO of Citius Pharmaceuticals. "We intend to study these cells in the clinic later this year to determine safety, efficacy, and the optimal dose of these cells in moderate to severe ARDS patients with COVID-19."

Citius has submitted a pre-IND meeting request and supporting briefing documents to the Center for Biologics Evaluation and Research ("CBER") of the FDA under the Coronavirus Treatment Acceleration Program (CTAP) for use of these MSCs for patients with Acute Respiratory Distress Syndrome (ARDS) due to SARS-CoV-2 disease.

Presentation Information:Title:Mesenchymal Stem Cells (MSCs) Generated Using mRNA Reprogramming Show Enhanced Growth Potential, Secretome, and Therapeutic Efficacy in a Demyelinating Disease ModelPresenter:Harris, Jasmine, Novellus, Inc.Date and Time: Wednesday, May 13 | 5:30 PM - 6:30 PM

About Acute Respiratory Distress Syndrome (ARDS)ARDS is a type of respiratory failure characterized by rapid onset of widespread inflammation in the lungs. ARDS is a rapidly progressive disease that occurs in critically ill patients most notably now in those diagnosed with COVID-19. ARDS affects approximately 200,000 patients per year in the U.S., exclusive of the current COVID-19 pandemic, and has a 30% to 50% mortality rate. ARDS is sometimes initially diagnosed as pneumonia or pulmonary edema (fluid in the lungs from heart disease). Symptoms of ARDS include shortness of breath, rapid breathing and heart rate, chest pain (particularly while inhaling), and bluish skin coloration. Among those who survive ARDS, a decreased quality of life is relatively common.

Story continues

About Coronavirus Treatment Acceleration Program (CTAP)In response to the pandemic, the FDA has created an emergency program called the Coronavirus Treatment Acceleration Program (CTAP) to accelerate the development of treatments for COVID-19. By redeploying staff, the FDA is responding to COVID-19-related requests and reviewing protocols within 24 hours of receipt. The FDA said CTAP "uses every available method to move new treatments to patients as quickly as possible, while at the same time finding out whether they are helpful or harmful." In practice, that means developers of potential treatments for COVID-19 will benefit from an unusually faster track at the FDA to shorten wait times at multiple steps of the process.

About Citius Pharmaceuticals, Inc.Citius is a late-stage specialty pharmaceutical company dedicated to the development and commercialization of critical care products, with a focus on anti-infectives and cancer care. For more information, please visit http://www.citiuspharma.com.

About Novellus, Inc.Novellus is a pre-clinical stage biotechnology company developing engineered cellular medicines using its non-immunogenic mRNA, nucleic-acid delivery, gene editing, and cell reprogramming technologies. Novellus is privately held and is headquartered in Cambridge, MA. For more information, please visit http://www.novellus-inc.com.

Safe HarborThis press release may contain "forward-looking statements" within the meaning of Section 27A of the Securities Act of 1933 and Section 21E of the Securities Exchange Act of 1934. Such statements are made based on our expectations and beliefs concerning future events impacting Citius. You can identify these statements by the fact that they use words such as "will," "anticipate," "estimate," "expect," "should," and "may" and other words and terms of similar meaning or use of future dates. Forward-looking statements are based on management's current expectations and are subject to risks and uncertainties that could negatively affect our business, operating results, financial condition, and stock price. Factors that could cause actual results to differ materially from those currently anticipated are: the risk of successfully negotiating a license agreement with Novellus within the option period; our need for substantial additional funds; the ability to access the FDA's CTAP program for the MARCO trial; the estimated markets for our product candidates, including those for ARDS, and the acceptance thereof by any market; risks associated with conducting trials for our product candidates, including those expected to be required for any treatment for ARDS and our Phase III trial for Mino-Lok; risks relating to the results of research and development activities; risks associated with developing our product candidates, including any licensed from Novellus, including that preclinical results may not be predictive of clinical results and our ability to file an IND for such candidates; uncertainties relating to preclinical and clinical testing; the early stage of products under development; risks related to our growth strategy; our ability to obtain, perform under, and maintain financing and strategic agreements and relationships; our ability to identify, acquire, close, and integrate product candidates and companies successfully and on a timely basis; our ability to attract, integrate, and retain key personnel; government regulation; patent and intellectual property matters; competition; as well as other risks described in our SEC filings. We expressly disclaim any obligation or undertaking to release publicly any updates or revisions to any forward-looking statements contained herein to reflect any change in our expectations or any changes in events, conditions, or circumstances on which any such statement is based, except as required by law.

Contact:Andrew ScottVice President, Corporate Development(O) 908-967-6677 x105 ascott@citiuspharma.com

View original content:http://www.prnewswire.com/news-releases/citius-announces-data-on-novecite-mesenchymal-stem-cells-nc-mscs-to-be-presented-at-the-american-society-of-gene-and-cell-therapy-asgct-annual-meeting-301058260.html

SOURCE Citius Pharmaceuticals, Inc.

See the original post here:
Citius Announces Data on NoveCite Mesenchymal Stem Cells (NC-MSCs) to be Presented at the American Society of Gene and Cell Therapy (ASGCT) Annual...

Novel CAR NK-cell technology could lead to new treatments for lupus, other incurable diseases – Stockhouse

CINCINNATI, May 12, 2020 The explosion in cellular immunotherapy that has revolutionized cancer care in recent years may soon begin showing potential application in treatment for lupus and other autoimmune diseases, thanks to a laboratory breakthrough led by experts at Cincinnati Children's and published in the journal Cell Reports Medicine.

In cancer, CAR T-cell therapy involves engineering T cells with chimeric antigen receptors (CAR) that allow them to recognize specific molecules on the surface of tumor cells. For certain forms of leukemia, some lung cancers, and other malignancies, this form of cellular immunotherapy has been life-changing for patients. Recent evidence suggests engineering natural killer (NK) cells to express CAR may be equally effective as T cells but with increased safety and clinical feasibility.

There is growing interest in the safety and efficacy of applying CAR cellular therapies to deadly and incurable autoimmune diseases. However, finding specific cell targets for diseases such as lupus has been much more difficultuntil now.

In a first-of-its-kind discovery, a team of Cincinnati Children's scientists led by Seth Reighard, PhD, Stephen Waggoner, PhD, and Hermine Brunner, MD, MSc, MBA, has engineered a CAR with the potential to revolutionize care of patients with lupus. When expressed by human NK cells, this CAR enables targeted elimination of T follicular helper (TFH) cells without harming other types of T-cells.

This treatment showed specificity in human cells in lab tests, and improved disease measures in a humanized mouse model of lupustwo key early signs of progress that suggest further research is warranted.

"This is the first method to specifically remove an otherwise intractable population of harmful cells," Waggoner says. "We think targeting them will be safe and clinically beneficial in multiple diseases. Our approach started with lupus because the disease is a leading cause of death in young women for which a cure is presently lacking."

The study appears in the first issue of the new, open access journal Cell Reports Medicine, which also carries a commentary about this new approach from Cecile King, PhD, an immunology expert at the Garvan Institute of Medical Research in Australia.

"Dysregulated TFH cells are associated with the development and severity of several autoimmune diseases and T cell malignancies," King states. "Indeed, the central role of TFH cells in many diseases has made them a major target for therapeutic modulation. The study by Reighard et al. provides exciting proof-of-principle evidence for the use of CAR NK cells in TFH-driven diseases."

How do CAR NK cells work?

The lupus-driving cells that the team wanted to eliminate lack cell-surface targets unique enough to distinguish them from other, desirable cells. To achieve selective targeting, the team realized a cardinal feature of TFH cells that could be exploited by carefully engineering the biochemistry of the CAR molecule.

Specifically, TFH express much greater quantities of a surface receptor, programmed cell death protein 1 (PD-1), than other cells that also express this receptor. Since activation of a CAR expressing NK cell is dependent on the strength of interaction between the CAR and its target receptor, as well as the number of such interactions between an NK cell and a target, the team engineered a CAR with relatively weak binding to PD-1. As a result, only cells like TFH that exhibit high expression of PD-1 trigger activation of the CAR NK and are eliminated as a result, which cells with lower levels of PD-1, including regulatory T cell (Treg) and memory T cells, are spared.

These programmed killer cells show early signs of potential as a therapy for systemic lupus erythematosus (SLE), which affects 20-150 per 100,000 people in the U.S. In fact, lupus ranks in the five causes of death among African American and Hispanic women, aged 15-34.

In addition, aberrant TFH responses play roles in several other autoimmune diseases, including Sjgren syndrome, juvenile dermatomyositis, multiple sclerosis, type 1 diabetes, and rheumatoid arthritis.

Although the potential toxicity of selectively eliminating TFH remains unexplored, the preservation of naive and memory CD4 T cells as well as B cells and other types of immune cells suggests that the state of immunodeficiency induced by these CAR NK cells will be far less severe than other immunotherapeutic strategies applied to autoimmune disease (e.g., rituximab).

Discovery based on years of research

This advance in CAR technology build upon previous work by Waggoner and colleagues in 2015 and 2018 that revealed how NK cells play surprising regulatory roles in infection and autoimmune disease.

The conceptual connections between infections and autoimmune diseases were further strengthened by a discovery led by John Harley, MD, PhD, and colleagues at Cincinnati Children's. In a 2018 study in Nature Genetics, they revealed how the Epstein-Barr virus uses groups of transcription factors to alter human DNA in ways that can increase a person's risk of developing lupus, multiple sclerosis, type 1 diabetes, and other diseases.

What's Next?

More work is needed to determine how much benefit can be gained by disrupting the role of TFH cells in lupus and other conditions. Concerns to address also include how to prevent the killer cells from attacking non-targeted "good" cells, and how to efficiently deliver the therapeutic cells.

Researchers are working to develop "suicide switches" for CAR NK cells that would make them safer for clinical use, Waggoner says. But importantly, NK cells appear to pose lower toxicity risk than CAR T-cell therapies in multiple clinical trials in cancer patients. Given the contributions of T cells to disease pathogenesis in lupus and other autoimmune disease, therapeutic NK cells likely yield additional benefit in these contexts.

Although the present study was performed with a human NK-cell line approved for clinical use by the FDA, the team envisions flexibility in the clinical application of the new CAR to lupus. CAR engineering of patient cells or cells from unrelated donors, including cord blood or induced pluripotent stem cell-derived NK cells, have all demonstrated excellent safety profiles while maintaining desirable efficacy in clinical trials.

"The CAR can be introduced to various effector cells using mRNA transfection, transposons, or viral vectors, Waggoner says. "Freezers full of CAR-expressing induced pluripotent stem cell-derived NK cells would provide an off-the-shelf product that could be rapidly and repeatedly administered to numerous patients in order to quell harmful flares of disease activity and promote sustained disease remission.

"If successes continue, a clinical trial might be possible within the next few years," Waggoner says.

View original content to download multimedia:http://www.prnewswire.com/news-releases/novel-car-nk-cell-technology-could-lead-to-new-treatments-for-lupus-other-incurable-diseases-301057216.html

SOURCE Cincinnati Children's Hospital Medical Center

Read more from the original source:
Novel CAR NK-cell technology could lead to new treatments for lupus, other incurable diseases - Stockhouse

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

Little Skates Could Hold the Key to Cartilage Therapy in Humans – Technology Networks

Nearly a quarter of Americans suffer from arthritis, most commonly due to the wear and tear of the cartilage that protects the joints. As we age, or get injured, we have no way to grow new cartilage. Unlike humans and other mammals, the skeletons of sharks, skates, and rays are made entirely of cartilage and they continue to grow that cartilage throughout adulthood.

And new research published this week in eLife finds that adult skates go one step further than cartilage growth: They can also spontaneously repair injured cartilage. This is the first known example of adult cartilage repair in a research organism. The team also found that newly healed skate cartilage did not form scar tissue.

"Skates and humans use a lot of the same genes to make cartilage. Conceivably, if skates are able to make cartilage as adults, we should be able to also," says Andrew Gillis, senior author on the study and a Marine Biological Laboratory Whitman Center Scientist from the University of Cambridge, U.K.

The researchers carried out a series of experiments on little skates (Leucoraja erinacea) and found that adult skates have a specialized type of progenitor cell to create new cartilage. They were able to label these cells, trace their descendants, and show that they give rise to new cartilage in an adult skeleton.

Why is this important? There are few therapies for repairing cartilage in humans and those that exist have severe limitations. As humans develop, almost all of our cartilage eventually turns into bone. The stem cell therapies used in cartilage repair face the same issue--the cells often continue to differentiate until they become bone. They do not stop as cartilage. But in skates, the stem cells do not create cartilage as a steppingstone; it is the end result.

"We're looking at the genetics of how they make cartilage, not as an intermediate point on the way to bone, but as a final product," says Gillis.

The research is in its early stages, but Gillis and his team hope that by understanding what genes are active in adult skates during cartilage repair, they could better understand how to stop human stem-cell therapies from differentiating to bone.

Note: There is no scientific evidence that "shark cartilage tablets" currently marketed as supplements confer any health benefits, including relief of joint pain.

Reference:Marconi, A., Hancock-Ronemus, A., & Gillis, J. A. (2020). Adult chondrogenesis and spontaneous cartilage repair in the skate, Leucoraja erinacea. ELife, 9. doi:10.7554/elife.53414

This article has been republished from the following materials. Note: material may have been edited for length and content. For further information, please contact the cited source.

See the original post:
Little Skates Could Hold the Key to Cartilage Therapy in Humans - Technology Networks