Sunday, May 20, 2018

PKD Research: Robots Grow Kidney Organoids from Stem Cells; Tweaking Cilia with STRIP

PKD Research

From University of Washington, Press Release

Robots grow mini-organs from human stem cells

microwell plate containing kidney organoids

Bird's eye view of a microwell plate containing kidney organoids, generated by liquid handling robots from human stem cells. Yellow boxed region is shown at higher magnification. Red, green, and yellow colors mark distinct segments of the kidney.


An automated system that uses robots has been designed to rapidly produce human mini-organs derived from stem cells. Researchers at the University of Washington School of Medicine in Seattle developed the new system.

The advance promises to greatly expand the use of mini-organs in basic research and drug discovery, according to Benjamin Freedman, assistant professor of medicine, Division of Nephrology, at the UW School of Medicine, who led the research effort.

"This is a new 'secret weapon' in our fight against disease,' said Freedman, who is a scientist at the UW Institute for Stem Cell and Regenerative Medicine, as well as at the Kidney Research Institute, a collaboration between the Northwest Kidney Centers and UW Medicine.

A report describing the new technique will be published online May 17 in the journal Cell Stem Cell. The lead authors were research scientists Stefan Czerniecki, and Nelly Cruz from the Freedman lab, and Dr. Jennifer Harder, assistant professor of internal medicine, Division of Nephrology at the University of Michigan School of Medicine, where she is a kidney disease specialist.

The traditional way to grow cells for biomedical research, Freedman explained, is to culture them as flat, two-dimensional sheets, which are overly simplistic. In recent years, researchers have been increasingly successful in growing stem cells into more complex, three-dimensional structures called mini-organs or organoids. These resemble rudimentary organs and in many ways behave similarly. While these properties make organoids ideal for biomedical research, they also pose a challenge for mass production. The ability to mass produce organoids is the most exciting potential applications of the new robotic technology, according to the developers.

In the new study, the researchers used a robotic system to automate the procedure for growing stem cells into organoids. Although similar approaches have been successful with adult stem cells, this is the first report of successfully automating the manufacture of organoids from pluripotent stem cells. That cell type is versatile and capable of becoming any type of organ.

In this process, the liquid-handling robots introduced the stem cells into plates that contained as many as 384 miniature wells each, and then coaxed them to turn into kidney organoids over 21 days. Each little microwell typically contained ten or more organoids, and each plate contained thousands of organoids. With a speed that would have impressed Henry Ford's car assembly line, the robots could produce many plates in a fraction of the time.

"Ordinarily, just setting up an experiment of this magnitude would take a researcher all day, while the robot can do it in 20 minutes," said Freedman.

"On top of that, the robot doesn't get tired and make mistakes," he added. "There's no question. For repetitive, tedious tasks like this, robots do a better job than humans."

The researchers further trained robots to process and analyze the organoids they produced. Harder and her colleagues at the University of Michigan Kidney Center used an automated, cutting-edge technique called single cell RNA sequencing to identify all the different types of cells found in the organoids.

"We established that these organoids do resemble developing kidneys, but also that they contain non-kidney cells that had not previously been characterized in these cultures," said Harder.

"These findings give us a better idea of the nature of these organoids and provide a baseline from which we can make improvements," Freedman said. "The value of this high-throughput platform is that we can now alter our procedure at any point, in many different ways, and quickly see which of these changes produces a better result."

Demonstrating this, the researchers discovered a way to greatly expand the number of blood vessel cells in their organoids to make them more like real kidneys.

The researchers also used their new technique to search for drugs that could affect disease. In one of these experiments, they produced organoids with mutations that cause polycystic kidney disease, a common, inherited condition that affects one in 600 people worldwide and often leads to kidney failure.
In this disease, tiny tubes in the kidneys and other organs swell like balloons and form expanding cysts that crowd out the healthy tissue.

In their experiment, the researchers exposed the polycystic kidney disease organoids to a number of substances. They found that one, a factor called blebbistatin that blocks a protein called myosin, led to a significant increase in the number and size of cysts.

"This was unexpected, since myosin was not known to be involved in PKD," Freedman said. Myosin, which is better known for its role in muscle contraction, may allow kidney tubules to expand and contract. If it is not functioning properly it might lead to cysts, Freedman explained.

"It's definitely a pathway we will be looking at," he said.

The title of the research paper in Cell Stem Cell is, "High-throughput screening enhances kidney organoid differentiation from human pluripotent stem cells and enables automated multidimensional phenotyping."

The research was supported by the American Society of Nephrology, PKD Foundation, National Kidney Foundation, Northwest Kidney Centers, Howard Hughes Medical Institute and the U.S. National Institutes of Health. The NIH grant numbers are: K01DK102826, UH3TR000504, UG3TR002158, K08DK089119, U54DK083912, R00DK094873, and R01DK097598.





From Eureka Alert

Scientists develop method to tweak tiny 'antenna' on cells

IMAGE


IMAGE: THESE ARE PRIMARY CILIA IN A MOUSE EMBRYONIC NODE. view more

CREDIT: SHINOHARA KYOSUKE, TOKYO UNIVERSITY OF AGRICULTURE AND TECHNOLOGY

Scientists at Johns Hopkins Medicine and the National Tsing Hua University in Taiwan say they have found a fast way to manipulate a cell's cilia, the tiny, fingerlike protrusions that "feel" and sense their microscopic environment. The experiments, performed in mouse cells, may advance scientists' efforts to not only understand how the nanosized antennae work, but also how to repair them.

A report of their findings appeared online April 30 in Nature Communications.

With few exceptions, most cells in the body have cilia or can grow them. The tiny antenna sense chemicals such as hormones and growth factors, which regulate cell health and growth. Cilia also detect mechanical and physical cues in the body, such as light, gravity, sound and the flow of blood and urine.

When cilia malfunction, a range of human diseases and conditions can occur. For example, problems with cilia in kidney cells can cause polycystic kidney disease, an incurable condition in which fluid-filled cysts interfere with kidney function and which is conventionally treated with dialysis.

Because cilia are so small -- 10,000 times smaller than a cell -- scientists have long found it challenging to squeeze their tools into such tight spaces to study them.

"When I was a postdoc, a colleague in a neighboring laboratory was studying cilia, and I hoped that by combining his knowledge of the biology of cilia with my expertise in cellular engineering, we could figure out how to manipulate cilia within their tiny spaces," says Takanari Inoue, Ph.D., professor of cell biology at the Johns Hopkins University School of Medicine and an author of the new report.

After years of work, he says they figured out a way to manipulate a chemical signaling pathway within cilia that controls how molecules are shuttled up and down the length of the tiny structure.

To do it, Inoue and his colleagues in Taiwan used a tool called chemically inducible dimerization, which they say is faster than efforts to manipulate the pathway by rewriting the cilia's genetic code. The tool, essentially, is a matchmaker -- it helps to mesh two specific chemicals together at specific sites within a living cell.

For the new study, Inoue and his colleagues added a protein called FRB to cells from mice grown in the laboratory. The FRB protein is capable of glomming onto a rigid structure within cilia, called a microtubule, which acts as a railway, shuttling proteins up and down the length of cilia.

Then they added a molecule called FKBP to the cells, which is attached to an enzyme that acts as an eraser for a chemical modification in the cilia called glutamylation. The FKBP and enzyme pair floats around the cell until scientists add a chemical called rapamycin, which causes FKBP to get trapped at FRB molecules within the cilia.

Once inside the cilia, the enzyme attached to the FKBP molecule selectively erases the glutamylation modification inside the cilia. It also ignores other signaling pathways.

The scientists call their molecule matchmaking STRIP, for spatiotemporal rewriting intraciliary post-translational modifications.

As a result of rapidly removing glutamylation in cilia, the scientists found that molecules flowed up the cilia, toward the tip, more slowly -- about .3 micrometers per second -- compared with .4 micrometers per second, using a dead enzyme that doesn't affect glutamylation.

"We think our technique is faster than existing means of tracking cilia activity and enables scientists to access cilia parts faster and dive into specific chemical modifications for certain amounts of time," says Inoue.

"Our STRIP system offers a new strategy for precisely controlling microtubule modifications in living cells. With this approach, it becomes possible to understand how microtubules regulate cellular functions and may also serve as a new way to treat human diseases in the future," says Yu-Chun Lin, Ph.D., an assistant professor at the Institute of Molecular Medicine at the National Tsing Hua University in Taiwan.

Other diseases affected by flawed cilia include a brain disorder called Joubert syndrome, a kidney disorder called nephronophthisis, retinitis pigmentosa and a rare disorder called situs inversus, in which the internal organs of the body are in the reverse location of their normal position.

The scientists also found that microtubules in the mouse cells that are not located inside cilia were not affected when they tinkered with glutamylation.

Inoue and his colleagues also found that the genetic output of a developmental pathway called Hedgehog (which is connected to glutamylation) is decreased in cells treated with STRIP compared with their controls.

Inoue and his colleagues say they now plan to apply STRIP to human cells and look more closely at the molecular process of glutamylation in cilia. They may also use STRIP to control other chemical modifications within cilia.

Sunday, May 13, 2018

ADPKD Treatment: FDA Approves ELISA Study: Phase 2 Clinical Trials for Lixivaptan: Regulus to conduct Phase 1 Multiple Ascending Dose Study

PKD Treatment Study

From Business Wire

Palladio Biosciences Receives FDA IND Clearance to Begin the ELISA Study, a Phase 2 Clinical Trial with Lixivaptan in Patients with Autosomal Dominant Polycystic Kidney Disease (ADPKD)


Palladio Biosciences, Inc. (Palladio) http://palladiobio.com/, a privately held biopharmaceutical company founded to develop medicines that make a meaningful impact on the lives of patients with orphan diseases of the kidney, today announces that the US Food and Drug Administration (FDA) has granted Palladio Biosciences Investigational New Drug (IND) clearance to proceed with a Phase 2 clinical trial of lixivaptan capsules in patients with autosomal dominant polycystic kidney disease (ADPKD).

The ELISA study is expected to begin enrolling patients at the end of June 2018 and is an open-label study which will enroll patients at several sites in the United States. It will pave the way for the initiation of a Phase 3 registration study in the first half of 2019.The ELISA Phase 2 study (Evaluation of Lixivaptan In Subjects with ADPKD), will evaluate the safety, pharmacokinetics and pharmacodynamics of multiple doses of lixivaptan in patients with ADPKD with relatively preserved kidney function (chronic kidney disease stages CKD1 and CKD2) and moderately impaired renal function (CKD3).

“We are very pleased that the FDA granted clearance of our Phase 2 trial of lixivaptan for patients with ADPKD” said Lorenzo Pellegrini, CEO of Palladio. “This is a pivotal event for our company as it marks the rebirth of lixivaptan as a clinical stage program for a disease with significant unmet medical need. We would like to take this opportunity to thank our advisors and collaborators for helping us meet this important milestone.”

“We are looking forward to advancing lixivaptan’s development program to provide a meaningfully differentiated treatment option for a broad population of ADPKD patients,” added Frank Condella, Palladio Biosciences’ Director. “We remain committed to working with patients, physicians and the PKD Foundation, the only organization in the U.S. solely dedicated to finding treatments and a cure for Polycystic Kidney Disease, to advance new treatments that improve the lives of patients with kidney disease.”

About Lixivaptan:

Lixivaptan was granted orphan designation by FDA for the treatment of ADPKD. It is a potent, selective vasopressin V2 receptor antagonist, a mechanism of action that has clinical proof of concept to slow kidney function decline in adults at risk of rapidly progressing ADPKD. Lixivaptan was previously administered to more than 1,600 subjects across 36 clinical studies as part of a prior clinical development program for the treatment of hyponatremia. Palladio expects to leverage lixivaptan’s large body of data generated in the hyponatremia clinical program to accelerate the development of lixivaptan for the treatment of ADPKD.

About Polycystic Kidney Disease (PKD) – Key Facts and Figures:

PKD is an inherited genetic disease that affects thousands of people in the United States and millions globally. ADPKD is the most common type of PKD. A person with ADPKD has a 50 percent chance of passing the disease on to each of his or her children. The disease is characterized by uncontrolled growth of fluid-filled cysts in the kidney, which can each grow to be as large as a football. Symptoms often include kidney infections and chronic pain. The continued enlargement of cysts and replacement of normal kidney tissue causes irreversible loss of renal function. In the United States, approximately 2,500 new people with PKD require dialysis or a kidney transplant every year, making PKD the 4th leading cause of kidney failure. There is no cure for PKD.




From Market Insider

Regulus Initiates Multiple Ascending Dose Study in Healthy Volunteers of RGLS4326 for the Treatment of ADPKD

Regulus Therapeutics Inc. (Nasdaq: RGLS), a biopharmaceutical company leading the discovery and development of innovative medicines targeting microRNAs, today announced that it has initiated a Phase I multiple ascending dose (MAD) study in healthy volunteers for RGLS4326 for the treatment of autosomal dominant polycystic kidney disease, or ADPKD. RGLS4326 is a novel, first-in-class, oligonucleotide designed to inhibit miR-17 using a unique chemistry that preferentially delivers to the kidney.

"We are pleased to advance the RGLS4326 program with the initiation of the MAD study, which will allow us to further characterize the safety and pharmacokinetic profile of RGLS4326 and establish the dose range that we will study in patients with ADPKD," said Timothy Wright, M.D., Chief R&D Officer of Regulus. "RGLS4326 represents a novel approach to treating ADPKD, a genetic disease leading to progressive loss of kidney function and kidney failure in the majority of patients."

This study is designed to characterize the safety, tolerability, pharmacokinetics and pharmacodynamics of increasing doses of RGLS4326. The MAD study was initiated based on data from the single ascending dose (SAD) Phase I study, which has completed dose escalation and continues in the planned follow-up phase. Preclinical studies with RGLS4326 have demonstrated a reduction in kidney cyst formation, improved kidney weight/body weight ratio, and decreased cyst cell proliferation in mouse models of ADPKD; and decreased cyst formation in cultured human ADPKD cyst cells in vitro.

About Autosomal Dominant Polycystic Kidney Disease (ADPKD)

ADPKD, caused by the mutations in the PKD1 or PKD2 genes, is among the most common human monogenetic disorders and a leading genetic cause of end-stage renal disease. The clinical hallmark of this disease is the development of multiple fluid filled cysts primarily in the kidneys and to a lesser extent in the liver and other organs. Excessive kidney tubule derived cyst cell proliferation, a central pathological feature, fuels the expansion of cysts, ultimately causing end-stage renal disease in approximately 50% of ADPKD patients by age 60. Approximately 1 in 1,000 people bear a mutation in either PKD1 or PKD2 genes worldwide.