"Sweet" Research Sheds Light on Glucose Metabolism in Neurons – Neuroscience News

Summary: Neurons in the brain directly metabolize glucose to function normally, contrary to previous beliefs that glial cells metabolized the sugar and indirectly fuel neurons. The findings could provide insights into the development of new therapeutic approaches for neurodegenerative diseases like Alzheimers and Parkinsons, where the brains uptake of glucose decreases in the early stages of the diseases.

Source: Gladstone Institute

The human brain has a sweet tooth, burning through nearly one quarter of the bodys sugar energy, or glucose, each day. Now, researchers at Gladstone Institutes and UC San Francisco (UCSF) have shed new light on exactly how neuronsthe cells that send electrical signals through the brainconsume and metabolize glucose, as well as how these cells adapt to glucose shortages.

Previously, scientists had suspected that much of the glucose used by the brain was metabolized by other brain cells called glia, which support the activity of neurons.

We already knew that the brain requires a lot of glucose, but it had been unclear how much neurons themselves rely on glucose and what methods they use to break the sugar down, saysKen Nakamura, MD, PhD,associate investigator at Gladstone and senior author of thenew study published in the journalCell Reports.Now, we have a much better understanding of the basic fuel that makes neurons run.

Past studies have established that the brains uptake of glucose is decreased in the early stages of neurodegenerative diseases like Alzheimers and Parkinsons. The new findings could lead to the discovery of new therapeutic approaches for those diseases and contribute to a better understanding of how to keep the brain healthy as it ages.

Simple Sugar

Many foods we eat are broken down into glucose, which is stored in the liver and muscles, shuttled throughout the body, and metabolized by cells to power the chemical reactions that keep us alive.

Scientists have long debated what happens to glucose in the brain, and many have suggested that neurons themselves dont metabolize the sugar. They instead proposed that glial cells consume most of the glucose and then fuel neurons indirectly by passing them a metabolic product of glucose called lactate. However, the evidence to support this theory has been scantin part because of how hard it is for scientists to generate cultures of neurons in the lab that do not also contain glial cells.

Nakamuras group solved this problem using induced pluripotent stem cells (iPS cells) to generate pure human neurons. IPS cell technology allows scientists to transform adult cells collected from blood or skin samples into any cell type in the body.

Then, the researchers mixed the neurons with a labeled form of glucose that they could track, even as it was broken down. This experiment revealed that neurons themselves were capable of taking up the glucose and of processing it into smaller metabolites.

To determine exactly how neurons were using the products of metabolized glucose, the team removed two key proteins from the cells using CRISPR gene editing. One of the proteins enables neurons to import glucose, and the other is required for glycolysis, the main pathway by which cells typically metabolize glucose. Removing either of these proteins stopped the breakdown of glucose in the isolated human neurons.

This is the most direct and clearest evidence yet that neurons are metabolizing glucose through glycolysis and that they need this fuel to maintain normal energy levels, says Nakamura, who is also an associate professor in the Department of neurology at UCSF.

Fueling Learning and Memory

Nakamuras group next turned to mice to study the importance of neuronal glucose metabolism in living animals. They engineered the animals neurons but not other brain cell typesto lack the proteins required for glucose import and glycolysis. As a result, the mice developed severe learning and memory problems as they aged.

This suggests that neurons are not only capable of metabolizing glucose, but also rely on glycolysis for normal functioning, Nakamura explains.

Interestingly, some of the deficits we saw in mice with impaired glycolysis varied between males and females, he adds. More research is needed to understand exactly why that is.

Myriam M. Chaumeil, PhD,associate professor at UCSF and co-corresponding author of the new work, has been developing specialized neuroimaging approaches, based on a new technology called hyperpolarized carbon-13, that reveal the levels of certain molecular products. Her groups imaging showed how the metabolism of the mices brains changed when glycolysis was blocked in neurons.

Such neuroimaging methods provide unprecedented information on brain metabolism, says Chaumeil. The promise of metabolic imaging to inform fundamental biology and improve clinical care is immense; a lot remains to be explored.

The imaging results helped prove that neurons metabolize glucose through glycolysis in living animals. They also showed the potential of Chaumeils imaging approach for studying how glucose metabolism changes in humans with diseases like Alzheimers and Parkinsons.

Finally, Nakamura and his collaborators probed how neurons adapt when they are not able to get energy through glycolysisas might be the case in certain brain diseases.

It turned out neurons use other energy sources, such as the related sugar molecule galactose. However, the researchers found that galactose was not as efficient a source of energy as glucose and that it could not fully compensate for the loss of glucose metabolism.

The studies we have carried out set the stage for better understanding how glucose metabolism changes and contributes to disease, says Nakamura.

His lab is planning future studies on how neuronal glucose metabolism changes with neurodegenerative diseases in collaboration with Chaumeils team, and how energy-based therapies could target the brain to boost neuronal function.

The first authors are Huihui Li and Yoshitaka Sei of Gladstone and Caroline Guglielmetti of UCSF. Other authors are Misha Zilberter, Lauren Shields, Joyce Yang, Kevin Nguyen, Neal Bennett, Iris Lo, and Yadong Huang of Gladstone; Lydia M. Le Page, Brice Tiret, Xiao Gao, and Martin Kampmann of UCSF; Talya L. Dayton and Matthew Vander Heiden of Massachusetts Institute of Technology; and Jeffrey C. Rathmell of Vanderbilt University Medical Center.

Funding: The work was supported by the National Institutes of Health (RF1 AG064170, R01 AG065428, AG065428-03S1, R01 NS102156, R21 AI153749 and RR18928), National Institute on Aging (R01 AG061150, R01 AG071697, P01 AG073082, R01 CA168653, R35 CA242379, R01 DK105550), the UCSF Bakar Aging Research Institute, the Alzheimers Association, a Bright Focus Foundation Award, a Berkelhammer Award for Excellence in Neuroscience, and a Chan Zuckerberg Initiative Neurodegeneration Challenge Network Ben Barres Early Career Acceleration Award.

Summary was written with the assistance of ChatGPT AI technology

Author: Julie LangelierSource: Gladstone InstituteContact: Julie Langelier Gladstone InstituteImage: The image is in the public domain

Original Research: Open access.Neurons require glucose uptake and glycolysis in vivo by Ken Nakamura et al. Cell Reports

Abstract

Neurons require glucose uptake and glycolysis in vivo

Neurons require large amounts of energy, but whether they can perform glycolysis or require glycolysis to maintain energy remains unclear. Using metabolomics, we show that human neurons do metabolize glucose through glycolysis and can rely on glycolysis to supply tricarboxylic acid (TCA) cycle metabolites.

To investigate the requirement for glycolysis, we generated mice with postnatal deletion of either the dominant neuronal glucose transporter (GLUT3cKO) or the neuronal-enriched pyruvate kinase isoform (PKM1cKO) in CA1 and other hippocampal neurons. GLUT3cKO and PKM1cKO mice show age-dependent learning and memory deficits.

Hyperpolarized magnetic resonance spectroscopic (MRS) imaging shows that female PKM1cKO mice have increased pyruvate-to-lactate conversion, whereas female GLUT3cKO mice have decreased conversion, body weight, and brain volume. GLUT3KO neurons also have decreased cytosolic glucose and ATP at nerve terminals, with spatial genomics and metabolomics revealing compensatory changes in mitochondrial bioenergetics and galactose metabolism.

Therefore, neurons metabolize glucose through glycolysisinvivoand require glycolysis for normal function.

Read this article:
"Sweet" Research Sheds Light on Glucose Metabolism in Neurons - Neuroscience News

Results of Study on Cryopreserved Hematopoietic Stem Cell Grafts … – GlobeNewswire

MINNEAPOLIS, April 18, 2023 (GLOBE NEWSWIRE) -- CIBMTR (Center for International Blood and Marrow Transplant Research) announced that the results of a multi-center observational study, around the impact of cryopreserved hematopoietic stem cell (HSC) grafts on patient survival rates were published in Blood Advances, a peer-reviewed open access medical journal published by the American Society of Hematology. The study showed that the shift in clinical practice to cryopreserved products necessitated during the pandemic did not adversely impact one-year overall survival. The CIBMTR is a research collaboration between the National Marrow Donor Program (NMDP)/Be The Match and the Medical College of Wisconsin (MCW).

The COVID-19 pandemic provided an unprecedented opportunity to study the impact of cryopreservation on clinical outcomes since the vast majority of patients received cryopreserved grafts for safety reasons at the onset of the pandemic. While it was comforting to find there were no differences in overall survival, there were more graft failures and relapses compared to fresh grafts, said Steven Devine, MD, Chief Medical Officer, NMDP/Be The Match and Senior Scientific Director, CIBMTR NMDP. These findings demonstrate that fresh grafts are preferred but that cryopreserved grafts do appear to be a good alternative during a crisis or if due to logistical reasons it could make the difference between transplant and no transplant.

The COVID-19 pandemic necessitated a substantial increase in the use of cryopreserved HSC grafts from both related and unrelated donors to ensure patients had a graft available prior to the start of conditioning for HCT. This cryopreservation necessitation was due to increased logistical challenges from international travel bans and fluctuating donor availability due to unpredictable health. However, pre-pandemic data on the impact of cryopreservation on post-transplant outcomes was limited. At the onset of the pandemic, the CIBMTR rapidly completed three retrospective analyses of outcomes in recipients of cryopreserved compared to fresh grafts administered prior to the pandemic with varying results and in all cases lack of a unifying rationale for use of cryopreservation.

The NMDP mandated cryopreservation of their facilitated collections at that onset of the pandemic and many centers adopted a similar approach for locally collected products. Thus, early in the pandemic the vast majority of patients received planned cryopreserved allografts allowing CIBMTR to successfully evaluate early post-HCT clinical outcomes in patients reported to the CIBMTR database who received a first allogeneic HCT using cryopreserved grafts. The study subjects were US patients receiving fresh (March-August 2019) or cryopreserved (March-August 2020) bone marrow or peripheral blood stem cell transplants from matched related or unrelated donors. This study included 1,543 and 2,499 recipients of cryopreserved and fresh products, respectively.

The results demonstrated that the shift in clinical practice to cryopreserved products necessitated during the pandemic did not adversely impact one-year post-transplant overall survival, non-relapse mortality, acute graft-versus-host disease (GVHD), or GVHD-free, relapse-free survival in recipients of cryopreserved versus fresh allografts. However, the study did find an adverse impact of cryopreservation on disease-free survival due to a higher risk of relapse. There was also an increased risk of primary graft failure following cryopreservation. One advantage observed with cryopreserved grafts was a decreased risk of chronic GVHD consistent with results previously described in a single center study published by Dana Farber Cancer Institute. Based on these results the study team concluded that fresh grafts are recommended, and that cryopreservation should be considered an option for patients when infusion of fresh grafts are not feasible.

NMDP/Be The Match and its research group CIBMTR are dedicated to providing clinical teams caring for HCT recipients with data that can inform their clinical practice, ensuring that patients thrive following transplant, said Amy Ronneberg, Chief Executive Officer, NMDP/ Be The Match. We are proud to have taken leadership on this important graft study and to have the results shared broadly in Blood Advances.

National Marrow Donor Program (NMDP)/Be The MatchThe National Marrow Donor Program (NMDP)/Be The Match is the leading global partner working to save lives through cellular therapy. With 35 years of experience managing the most diverse registry of potential unrelated blood stem cell donors and cord blood units in the world, NMDP/Be The Match is a proven partner in providing cures to patients with life-threatening blood and marrow cancers and diseases. Through their global network, they connect centers and patients to their best cell therapy optionfrom blood stem cell transplant to a next-generation therapyand collaborate with cell and gene therapy companies to support therapy development and delivery through Be The Match BioTherapies. NMDP/Be The Match is a tireless advocate for the cell therapy community, working with hematologists/oncologists to remove barriers to consultation and treatment, and supporting patients through no-cost programs to eliminate non-medical obstacles to cell therapy. In addition, they are a global leader in research through the CIBMTR (Center for International Blood and Marrow Transplant Research)a collaboration with Medical College of Wisconsin, investing in and managing research studies that improve patient outcomes and advance the future of care.

CIBMTR (Center for International Blood and Marrow Transplant Research)Center for International Blood and Marrow Transplant Research is a nonprofit research collaboration between the National Marrow Donor Program (NMDP)/ Be The Match, in Minneapolis, and the Medical College of Wisconsin, in Milwaukee. The CIBMTR collaborates with the global scientific community to increase survival and enrich quality of life for patients. CIBMTR facilitates critical observational and interventional research through scientific and statistical expertise, a large network of centers, and a unique database of long-term clinical data for more than 630,000 people who have received hematopoietic cell transplantation and other cellular therapies. Learn more at cibmtr.org.

Media Contacts

NMDP/Be The MatchClarity Quest, 877-887-7611 Bonnie Quintanilla, bonnie@clarityqst.comPhyllis Grabot, phyllis@clarityqst.com

Read the rest here:
Results of Study on Cryopreserved Hematopoietic Stem Cell Grafts ... - GlobeNewswire

Healing the unhealable: New approach helps bones mend themselves – Newswise

Newswise Young babies and newborn mice can naturally heal damage to the bones that form the top of the skull, but this ability is lost in adults. In a new study published inProceedings of the National Academy of Sciences, University of Pittsburgh researchers developed a novel approach that promoted bone regeneration in mice without implantation of bone tissue or biomaterials.

The technique uses a device similar to an orthodontic wire used to realign teeth to carefully stretch the skull along its sutures, activating skeletal stem cells that reside in these wiggly seams. In adult mice, the technique repaired damage to the skull that otherwise would not have healed on its own.

Our approach is inspired by babies because they have an amazing ability to regenerate bone defects in the calvarial bones that make up the top of the skull, said senior author Giuseppe Intini, D.D.S., Ph.D., associate professor of periodontics and preventive dentistry at thePitt School of Dental Medicine,member of theMcGowan Institute for Regenerative Medicineand an investigator atUPMC Hillman Cancer Center. By harnessing the bodys own healing capacity with autotherapies, we can stimulate bone to heal itself. We hope to build on this research in the future to develop novel therapies for people.

Trauma, congenital defects and surgery to treat cancer or other diseases are common causes of damage to the skull. After people reach the age of about 2 years, such injuries dont heal on their own.

In babies, the calvarial bones are not completely fused, so the sutures where stem cells reside are still open, said Intini. We wondered whether the unfused sutures had something to do with the bone regenerative capacity observed in babies and hypothesized that we could reverse engineer this in adults by mechanically opening the sutures to activate the stem cell niche and boost stem cell numbers.

In mice which have very similar skull development to humans the researchers used a so-called bone distraction device to carefully apply a controlled pulling force to the calvarial bones, strong enough to slightly widen the sutures but not enough to cause a fracture. Using single-cell RNA sequencing and live-imaging microscopy, they found that the number of stem cells in the expanded sutures of these animals quadrupled.

As a result, mice treated with the device regenerated bone to heal a large defect in the skull.

If you can effectively activate the stem cell niche, you can increase the number of stem cells and sustain regeneration of bone defects, said Intini. Remarkably, we showed that the defect can heal even if its away from the suture.

Although the approach was effective in healing skeletally mature 2-month-old mice, the age that roughly translates to young adulthood in humans, it did not work in 10-month-old, or middle-aged, rodents.

In older mice, the quantity of stem cells in calvarial sutures is very low, so expanding this niche is not as effective in boosting healing capacity, Intini explained. Overcoming this challenge is a focus of research to come.

Current treatments for damage to the skull are usually bone grafts or implantation of biomaterials that act as scaffolds for bone regeneration, but these approaches are not always effective and come with risks, said Intini.

The researchers are investigating how their findings could be used to inform novel therapies in people, not just to heal skull injuries but also fractures in long bones such as the femur. Bone distraction devices are already used to treat certain conditions such as a birth defect called craniosynostosis, in which the calvarial bones fuse too early, so expanding this technique to promote bone regeneration could be a future focus of clinical trials.

Intini and his team are also investigating non-mechanical approaches to activate skeletal stem cells such as medications.

Other authors who contributed to the study were Zahra A. Aldawood, D.M.Sc, of the Harvard School of Dental Medicine and Imam Abdulrahman Bin Faisal University; Luigi Mancinelli, Ph.D., Xuehui Geng, M.D., M.S., Taiana C. Leite, D.D.S., M.S., and Roberta Di Carlo, Ph.D., all of Pitt; Shu-Chi A. Yeh, Ph.D., and Charles P. Lin, Ph.D., both of Massachusetts General Hospital; Jonas Gustafson, of Seattle Childrens Research Institute; Katarzyna Wilk, M.S., Joseph Yozgatian, D.D.S., M.M.Sc., Ph.D., Sasan Garakani, D.D.S., and Seyed Hossein Bassir, D.D.S., D.M.Sc., of the Harvard School of Dental Medicine; and Michael L. Cunningham, M.D., Ph.D., of the Seattle Childrens Research Institute and the University of Washington.

This research was supported by the National Institutes of Healths National Institute of Dental and Craniofacial Research (grants #R00DE021069 and #R01DE026155).

Read the rest here:
Healing the unhealable: New approach helps bones mend themselves - Newswise

11 Incredible Animals That Regenerate – AZ Animals

There are countless natural wonders around the globe. They are too numerous to count, and we humans never get tired of the surprises nature has in store for us.

In addition to all the marvels, it is worthwhile to consider if we would be able to regrow a lost limb or damaged organ. These skills may seem like something out of a sci-fi film, but they are really found in the animal kingdom. Although the majority of animals lack these skills, there are some animals that regenerate limbs, organs, and other body parts with ease. These few organisms could help us understand how regeneration works in their species and possibly even ours. Who knows? Maybe science could use these fascinating creatures to one day make regeneration possible for humans. In fact, many of the animals on this list are currently being studied for their potential use in regenerative medicine for humans.

Pseudoscience aside, lets take a look at some real, living animals that regenerate! But first, lets ask a very important question: Why cant humans regenerate?

So how exactly can the species listed in this article regenerate? And why are the majority of creatures, including humans and other mammals, so terrible at regeneration? According to science, thats still a confounding topic today. There are a few competing theories, and the scientific community is still undecided.

One theory relates to how our immune system has evolved. Mammals and birds, which have very high immunity levels, cannot regrow their legs, fingers, and other body parts. This could be a result of the immune systems desire to avoid cancerous tumor growth and the fact that the molecular pathways of regeneration and tumor growth are identical, including the usage of stem cells. Therefore, evolution guarantees that these animals wont have as many cancers, but they also wont regenerate.

Research on the African spiny mouse, a species of mouse that can regrow its skin and hair after an injury, lends credence to this notion. According to a few studies, the skin these animals regenerate doesnt appear to include any immune cells called macrophages. Macrophages are white blood cells stimulated by the bodys immune system. Because of this, a large portion of the scientific community believes that immunity and regeneration are somehow related.

If and when humans are able to use any of these regeneration abilities will depend on advancements in our understanding of how and why certain animals can regenerate while others cannot. Doctors, scientists, and other professionals involved in the field of regenerative medicine should pay particular attention to this. Humans, for instance, cannot create new fingers or legs, but during fetal development, these genes all help the formation of our fingers or legs, and they are also present in starfish and hydra, which are regenerative animals mentioned later on in this guide.

Perhaps a method to activate these genes during postnatal development to restore limbs will emerge. Maybe mankind will find some way to make human regeneration possible. For now, though, its all still a pretty big mystery.

Now that we understand a bit more about regeneration in animals, lets take a look at a few animals that regenerate!

Classification: Asteroidea class

Sea stars have the capacity to regenerate their tube feet and ray arms after accidents. Most sea stars, also known as starfish, have five limbs, but some have as many as 40. Because the majority of their essential organs are located in their arms, certain sea stars can regenerate complete bodies or a new sea star merely from a section of a severed limb. When predators catch them, they can also release or drop one arm.

Starfish are capable of developing a new body from a lost limb in addition to a new limb. The original starfish can be broken apart into several new ones. Fission is a term used to describe this type of asexual reproduction. Fission occurs when the starfish loses one or more of its limbs and its central disc splits into two parts. From there, another sea star is created that is genetically identical to the parent plant.

iStock.com/Damocean

Classification: Ambystoma mexicanum

Axolotls are a type of aquatic salamander that have remarkable regeneration powers. They can grow new skin, limbs, organs, or just about any other part of the body. Axolotls live permanently underwater because they never develop lungs and instead retain their gills. Axolotls can regenerate limbs and organs flawlessly and without leaving any scarring, which is even more astounding. In as short as three weeks, they can repeat this as often as required.

The Axolotl is the only vertebrate, regardless of age, that can regenerate a number of its body parts. However, it does not use its stem cell population to do this. Instead, it takes advantage of a process called dedifferentiation. When their bodies are damaged, neighboring undifferentiated cells help them form a stub known as a blastema.

These animals basically turn back the clock on their bodys aged cells so they may begin to behave like embryonic or stem cells, despite the fact that they are not stem cells. They havent undergone differentiation since they fall midway between stem cells and adult cells, but they are already pre-programmed for what they will become. Many other creatures with the potential to regenerate prefer this method of regeneration, which is known as epimorphic regeneration. Salamanders and terrestrial lizards also employ this strategy. The starfish does as well, and occasionally it can develop a completely new body from just one arm.

Spok83/Shutterstock.com

Classification: Selachimorpha superorder

Now this is a surprising entry! Sharks can renew their dental structures, but they cannot restore their organs or other bodily components like other animals on this list can. Over the course of a lifetime, they lose at least 30,000 teeth. However, each one may regenerate in a matter of days or months. Over the course of its lifetime, a shark can regenerate missing teeth up to 50 times.

A sharks ability to regenerate teeth might take anywhere from a few days and several months. Dentistry could undergo a real revolution if scientists can figure out how this regeneration process works!

Alessandro De Maddalena/Shutterstock.com

Classification: Planaria genus

Flatworms known as planarians have a remarkable capacity for self-regeneration. In just a few weeks, one might create two planarians by slicing one in half; each half would quickly fill in the gaps in a very short amount of time.

One of the most remarkable regeneration techniques in the animal kingdom is used by these flatworms. These aquatic worms are invertebrates, and even after losing up to 90% of their bodies to damage, they can completely rebuild their whole bodies. They can even grow their head back if they are decapitated.

These creatures regenerate via a stem cell-mediated process. They have a population of pluripotent stem cells that are constantly present in the body and are intermittently replacing damaged cells. These cells are effectively tasked to repair the missing structure when a significant amputation occurs, no matter how severe. Sea squirts, which are a type of marine invertebrate, also employ this method.

Rattiya Thongdumhyu/Shutterstock.com

Classification: Urodela order

The salamander is an amphibian with short legs and a tail. The number of salamander species that we currently know of exceeds 700. Although all salamander species are capable of some degree of regeneration, certain species are more capable than others. Following the removal of the old tail to frighten away predators, certain salamanders can develop a new tail in a few weeks. The replacement limb performs all functions just like the old one.

Salamanders have earned praise for being masters of regeneration because of their astonishing capacity to create new tissues, organs, and even whole body parts, like their limbs. The methods by which salamander cells, tissues, and organs detect and restore missing or damaged pieces can provide key insights into the world of regenerative medicine.

iStock.com/Wirestock

Classification: Hydra genus

The hydra is a type of freshwater jellyfish that prefers to adhere to rocks throughout its life, similar to an anemone. These unique animals often go through a process of regeneration called morphallaxis.

In essence, these animals can shuffle their cells around and restructure whats left of the tissue, creating a miniature replica that is completely formed and has all of the necessary features. They can also take this regeneration technique a step further. The mechanism of how they regenerate can change depending on how they are harmed. If they sustain more severe wounds, the hydra will also engage in the same process as the Axolotl, whereby a fresh batch of cells proliferates and dedifferentiates to fill in the gaps in the missing structure.

Lebendkulturen.de/Shutterstock.com

Classification: Ascidiacea class

Tunicates, sometimes referred to as sea squirts, are renowned for their extraordinary ability to regenerate their whole body. A sea squirt can restructure its residual tissues and rebuild a completely functioning body in a couple of days after being damaged or losing a large chunk of its body.

Genes that regulate cell division and differentiation are activated during the regeneration process in sea squirts. In order to create the required tissues and organs, the cells must then rearrange and differentiate. Sea squirts are a model organism for researching the genetic and molecular pathways of regeneration because of their exceptional capacity for regeneration. This capacity could also provide new ideas for regenerative medicine, like many of the entries on this list.

Samuel Chow / Creative Commons

Classification: Slender danios

Even as older adults, zebrafish have the ability to regenerate their fins, spinal cord, retinas, heart, kidneys, and the telencephalon, the most advanced portion of the frontal lobe of the brain. It appears that different organs have different pathways for regeneration in this creature as well. The Axolotl or the starfish have comparable processes used for fin regeneration. However, just like the flatworm, regeneration of the zebrafishs telencephalon relies on stem cells to intervene and ensure the fishs brain is properly repaired.

Ian Grainger/Shutterstock.com

Classification: Astyanax mexicanus

Mexican tetras can repair heart tissue, much like zebrafish. Or rather, surface fish of this species can; populations of fish from caves no longer possess this ability. After damage, cave populations hearts develop scarring similar to how a humans heart would. According to the latest research on this species, tetras have unregulated versions of many genes.

The surface specimens of the Mexican tetra, which live in rivers and streams, can regenerate tissue without leaving scars. Researchers are hoping that their research on the Mexican tetra will help them make advances in the treatment of cardiovascular disease. The Mexican tetra is not the only fish capable of regenerating heart tissue, though. Also capable of regenerating its heart with minimal to no scarring is the zebrafish.

Kuttelvaserova Stuchelova/Shutterstock.com

Classification: Chamaeleonidae family

Chameleons are extremely fascinating creatures that are widely renowned for their extraordinary ability to alter their color in order to fit in with their surroundings. Chameleons can also grow new tails and limbs, in addition to their other abilities. During the healing process, they can also repair damaged skin and nerves.

A chameleon can sprout a new tail if it loses its original one. A blastema, or a collection of undifferentiated cells that will eventually become the new tail, is created throughout the process. Cells from the tail stump that dedifferentiate, or go back to a less specialized state, create the blastema. Following cell division and differentiation, the cells form the diverse tissues of the new tail. This remarkable capacity for regeneration is displayed by a few other species and is a subject of current study in the field of regenerative medicine.

Lauren Suryanata/Shutterstock.com

Classification: Cervidae family

Deer antlers are the only organ in mammals that can totally regenerate. They lose their antlers each year and then re-grow into enormous, branching structures of bone and cartilage that are utilized for combat and exhibition.

Scientists are using the regeneration of antlers, which is started and maintained by stem cells generated from the neural crest, to mimic and research the regeneration of other animal organs. Only male deer (except for caribou) have antlers. Male deer grow antlers in order to compete with other males for females and to find food in the snow. Antlers develop at a very rapid rate of roughly one-fourth of an inch every day.

Bob Keefer/Shutterstock.com

There are many animals that regenerate around the world. The animals weve listed above are just a few. Hopefully, one day science will be able to harness the processes of regeneration that these animals possess to apply to humans.

See the rest here:
11 Incredible Animals That Regenerate - AZ Animals

Reinforcement learning: From board games to protein design – EurekAlert

image:Examples of protein architectures designed through a software program that uses reinforcement learning. view more

Credit: Ian Haydon/ UW Medicine Institute for Protein Design

Scientists have successfully applied reinforcement learning to a challenge in molecular biology.

The team of researchersdeveloped powerful new protein design software adapted from a strategy proven adept at board games like Chess and Go. In one experiment, proteins made with the new approach were found to be more effective at generating useful antibodies in mice.

The findings, reported April 21 in Science, suggest that this breakthrough may soon lead to more potent vaccines. More broadly, the approach could lead to a new era in protein design.

"Our results show that reinforcement learning can do more than master board games. When trained to solve long-standing puzzles in protein science, the software excelled at creating useful molecules," said senior author David Baker, professor of biochemistry at the UW School of Medicine in Seattle and a recipient of the 2021 Breakthrough Prize in Life Sciences.

"If this method is applied to the right research problems, he said, it could accelerate progress in a variety of scientific fields."

The research is a milestone in tapping artificial intelligence to conduct protein science research. The potential applications are vast, from developing more effective cancer treatments to creating new biodegradable textiles.

Reinforcement learning is a type of machine learning in which a computer program learns to make decisions by trying different actions and receiving feedback. Such an algorithm can learn to play chess, for example, by testing millions of different moves that lead to victory or defeat on the board. The program is designed to learn from these experiences and become better at making decisions over time.

To make a reinforcement learning program for protein design, the scientists gave the computer millions of simple starting molecules. The software then made ten thousand attempts at randomly improving each toward a predefined goal. The computer lengthened the proteins or bent them in specific ways until it learned how to contort them into desired shapes.

Isaac D. Lutz, Shunzhi Wang, and ChristofferNorn, all members of the Baker Lab, led the research. Their teams Science manuscript is titled "Top-down design of protein architectures with reinforcement learning."

"Our approach is unique because we use reinforcement learning to solve the problem of creating protein shapes that fit together like pieces of a puzzle," explained co-lead author Lutz, a doctoral student at the UW Medicine Institute for Protein Design. "This simply was not possible using prior approaches and has the potential to transform the types of molecules we can build."

As part of this study, the scientists manufactured hundreds of AI-designed proteins in the lab. Using electron microscopes and other instruments, they confirmed that many of the protein shapes created by the computer were indeed realized in the lab.

This approach proved not only accurate but also highly customizable. For example, we asked the software to make spherical structures with no holes, small holes, or large holes. Its potential to make all kinds of architectures has yet to be fully explored, said co-lead author Shunzhi Wang, a postdoctoral scholar at the UW Medicine Institute for Protein Design.

The team concentrated on designing new nano-scale structures composed of many protein molecules. This required designing both the protein components themselves and the chemical interfaces that allow the nano-structures to self-assemble.

Electron microscopy confirmed that numerous AI-designed nano-structures were able to form in the lab. As a measure of how accurate the design software had become, the scientists observed many unique nano-structures in which every atom was found to be in the intended place. In other words, the deviation between the intended and realized nano-structure was on average less than the width of a single atom. This is called atomically accurate design.

The authors foresee a future in which this approach could enable them and others to create therapeutic proteins, vaccines, and other molecules that could not have been made using prior methods.

Researchers from the UW Medicine Institute for Stem Cell and Regenerative Medicine used primary cell models of blood vessel cells to show that the designed protein scaffolds outperformed previous versions of the technology. For example, because the receptors that help cells receive and interpret signals were clustered more densely on the more compact scaffolds, they were more effective at promoting blood vessel stability.

Hannele Ruohola-Baker, a UW School of Medicine professor of biochemistry and one of the studys authors, spoke to the implications of the investigation for regenerative medicine: The more accurate the technology becomes, the more it opens up potential applications, including vascular treatments for diabetes, brain injuries, strokes, and other cases where blood vessels are at risk. We can also imagine more precise delivery of factors that we use to differentiate stem cells into various cell types, giving us new ways to regulate the processes of cell development and aging.

This work was funded by the National Institutes of Health (P30 GM124169, S10OD018483, 1U19AG065156-01, T90 DE021984, 1P01AI167966); Open Philanthropy Project and The Audacious Project at the Institute for Protein Design; Novo Nordisk Foundation (NNF170C0030446); Microsoft; and Amgen. Research was in part conducted at the Advanced Light Source, a national user facility operated by Lawrence Berkeley National Laboratory on behalf of the Department of Energy

News release written by Ian Haydon, UW Medicine Institute for Protein Design.

Computational simulation/modeling

Not applicable

Top-down design of protein architectures with reinforcement learning

21-Apr-2023

David Baker, Shunzhi Wang, Isaac D. Lutz, Christoffer Norn, Annie Dosey, Neil P. King, and Andrew J. Borst are inventors on a provisional patent application (63/383,700) submitted by the University of Washington for the design, composition, and applications of the protein assemblies described in this work. The remaining authors declare no competing interests.

Disclaimer: AAAS and EurekAlert! are not responsible for the accuracy of news releases posted to EurekAlert! by contributing institutions or for the use of any information through the EurekAlert system.

Originally posted here:
Reinforcement learning: From board games to protein design - EurekAlert

Tonix Pharmaceuticals Announces Presentations of Pre-Clinical Data on TNX-1700 in Syngeneic Models of Colorectal and Gastric Cancer at the American…

Tonix Pharmaceuticals Holding Corp.

CHATHAM, N.J., April 19, 2023 (GLOBE NEWSWIRE) -- Tonix Pharmaceuticals Holding Corp. (Nasdaq: TNXP), a clinical-stage biopharmaceutical company, today announced the presentation of two posters with research results on TNX-1700 (recombinant TFF2 albumin fusion peptide) at the American Association for Cancer Research (AACR) Annual Meeting, held April 14-19, 2023, in Orlando, Fla. Copies of the Companys posters are available under the Scientific Presentations tab of the Tonix website at http://www.tonixpharma.com.

The poster presentation, titled, MDSC-targeted TFF2-MSA suppresses tumor growth and increases survival in anti-PD-1 treated MC38 and CT26.wt murine colorectal cancer models, includes data demonstrating that targeting myeloid-derived suppressor cells (MDSCs) using murine TNX-1700, or mTNX-1700 (TFF2-MSA fusion protein) synergizes with PD-1 blockade therapy in advanced syngeneic mouse models of colorectal cancer. The data show that mTNX-1700 and anti-PD-1 monotherapy each were able to evoke anti-tumor immunity in the MC38 and CT26.wt models of colorectal cancer, and that mTNX-1700 augmented the anti-tumor efficacy of anti-PD-1 therapy in both of these colorectal cancer models.

The poster presentation, titled, MDSC-targeted TFF2-MSA synergizes with PD-1 blockade therapy in diffuse-type gastric cancer, includes data showing that targeting MDSCs using mTNX-1700 synergizes with PD-1 blockade therapy in advanced and metastatic syngeneic mouse models of diffuse-type gastric cancer, suggesting combination therapy of mTNX-1700 and PD-1 blockade may also be applicable to gastric cancer.

We believe these data demonstrate that targeting MDSCs using mTNX-1700 provides additive benefits to PD-1 blockade therapy in advanced and metastatic syngeneic mouse models of colorectal and gastric cancer, said Seth Lederman, M.D., Chief Executive Officer of Tonix Pharmaceuticals.

About Trefoil Factor Family Member 2 (TFF2)

Human TFF2 is a secreted protein, encoded by the TFF2 gene in humans, that is expressed in gastrointestinal mucosa where it functions to protect and repair mucosa. TFF2 is also expressed at low levels in splenic immune cells and is now appreciated to have intravascular roles in the spleen and in the tumor microenvironment. In gastric cancer, TFF2 is epigenetically silenced, and TFF2 is suggested to be protective against cancer development through several mechanisms. Tonix is developing TNX-1700 (rTFF2-HSA) for the treatment of gastric and colon cancers under a license from Columbia University. The inventor at Columbia is Dr. Timothy Wang, who is an expert in the molecular mechanisms of carcinogenesis whose research has focused on the carcinogenic role of inflammation in modulating stem cell functions. Dr. Wang demonstrated that knocking out the mTFF2 gene in mice leads to faster tumor growth and that overexpression of TFF2 markedly suppresses tumor growth by curtailing the homing, differentiation, and expansion of MDSCs to allow activation of cancer-killing CD8+ T cells.1 He went on to show that a novel engineered form of recombinant murine TFF2 (mTFF2-CTP) had an extended half-lifein vivoand was able to suppress MDSCs and tumor growth in an animal model of colorectal cancer. Later, he showed in gastric cancer models that suppressing MDSCs using chemotherapy enhances the effectiveness of anti-PD1 therapy and significantly reduces tumor growth.2Dr. Wang proposed the concept of employing rTFF2 in combination with other therapies in cancer prevention and early treatment. Dr. Wang presented data at the American Association for Cancer Research (AACR) conference as a collaboration between Tonix and Columbia University in 2020that includes data from a preclinical study which investigated the role of PD-L1 in colorectal tumorigenesis and evaluated the utility of targeting myeloid-derived suppressor cells (MDSCs) in combination with PD-1 blockade in mouse models of colorectal cancer. The data show that anti-PD-1 monotherapy was unable to evoke anti-tumor immunity in this model of colorectal cancer, but mTFF2-CTP augmented the efficacy of anti-PD-1 therapy. Anti-PD-1 in combination with TFF2-CTP showed greater anti-tumor activity in PD-L1-overexpressing mice.

Story continues

Tonix Pharmaceuticals Holding Corp.*

Tonix is a clinical-stage biopharmaceutical company focused on discovering, licensing, acquiring and developing therapeutics to treat and prevent human disease and alleviate suffering. Tonixs portfolio is composed of central nervous system (CNS), rare disease, immunology and infectious disease product candidates. Tonixs CNS portfolio includes both small molecules and biologics to treat pain, neurologic, psychiatric and addiction conditions. Tonixs lead CNS candidate, TNX-102 SL (cyclobenzaprine HCl sublingual tablet), is in mid-Phase 3 development for the management of fibromyalgia with topline data expected in the fourth quarter of 2023. TNX-102 SL is also being developed to treat Long COVID, a chronic post-acute COVID-19 condition. Enrollment in a Phase 2 study has been completed, and topline results are expected in the third quarter of 2023. TNX-1900 (intranasal potentiated oxytocin), in development for chronic migraine, is currently enrolling with topline data expected in the fourth quarter of 2023. TNX-601 ER (tianeptine hemioxalate extended-release tablets), a once-daily formulation being developed as a treatment for major depressive disorder (MDD), is also currently enrolling with interim data expected in the fourth quarter of 2023. TNX-1300 (cocaine esterase) is a biologic designed to treat cocaine intoxication and has been granted Breakthrough Therapy designation by the FDA. A Phase 2 study of TNX-1300 is expected to be initiated in the second quarter of 2023. Tonixs rare disease portfolio includes TNX-2900 (intranasal potentiated oxytocin) for the treatment of Prader-Willi syndrome. TNX-2900 has been granted Orphan Drug designation by the FDA. Tonixs immunology portfolio includes biologics to address organ transplant rejection, autoimmunity and cancer, including TNX-1500, which is a humanized monoclonal antibody targeting CD40-ligand (CD40L or CD154) being developed for the prevention of allograft and xenograft rejection and for the treatment of autoimmune diseases. A Phase 1 study of TNX-1500 is expected to be initiated in the second quarter of 2023. Tonixs infectious disease pipeline includes TNX-801, a vaccine in development to prevent smallpox and mpox, for which a Phase 1 study is expected to be initiated in the second half of 2023. TNX-801 also serves as the live virus vaccine platform or recombinant pox vaccine platform for other infectious diseases. The infectious disease portfolio also includes TNX-3900 and TNX-4000, classes of broad-spectrum small molecule oral antivirals.

*All of Tonixs product candidates are investigational new drugs or biologics and have not been approved for any indication.1Dubeykovskaya ZA et al, Nat Commun 20162Kim W et al, Gastroenterology 2021

This press release and further information about Tonix can be found at http://www.tonixpharma.com.

Forward Looking Statements

Certain statements in this press release are forward-looking within the meaning of the Private Securities Litigation Reform Act of 1995. These statements may be identified by the use of forward-looking words such as anticipate, believe, forecast, estimate, expect, and intend, among others. These forward-looking statements are based on Tonix's current expectations and actual results could differ materially. There are a number of factors that could cause actual events to differ materially from those indicated by such forward-looking statements. These factors include, but are not limited to, risks related to the failure to obtain FDA clearances or approvals and noncompliance with FDA regulations; delays and uncertainties caused by the global COVID-19 pandemic; risks related to the timing and progress of clinical development of our product candidates; our need for additional financing; uncertainties of patent protection and litigation; uncertainties of government or third party payor reimbursement; limited research and development efforts and dependence upon third parties; and substantial competition. As with any pharmaceutical under development, there are significant risks in the development, regulatory approval and commercialization of new products. Tonix does not undertake an obligation to update or revise any forward-looking statement. Investors should read the risk factors set forth in the Annual Report on Form 10-K for the year ended December 31, 2022, as filed with the Securities and Exchange Commission (the SEC) on March 13, 2023, and periodic reports filed with the SEC on or after the date thereof. All of Tonix's forward-looking statements are expressly qualified by all such risk factors and other cautionary statements. The information set forth herein speaks only as of the date thereof.

Contacts

Jessica Morris (corporate)Tonix Pharmaceuticalsinvestor.relations@tonixpharma.com(862) 904-8182

Maddie Stabinski (media)Russo Partnersmadeline.stabinski@russopartnersllc.com (212) 845-4273

Peter Vozzo (investors)ICR Westwickepeter.vozzo@westwicke.com(443) 213-0505

Read more from the original source:
Tonix Pharmaceuticals Announces Presentations of Pre-Clinical Data on TNX-1700 in Syngeneic Models of Colorectal and Gastric Cancer at the American...

MNK proteins as therapeutic targets in leukemia | OTT – Dove Medical Press

Plain Language Summary

Identification and vetting of new targets in cancer medicine is essential for the development of treatments to improve survival of patients. MNKs are kinases involved in cancer promoting and cancer cell survival signaling. Preclinical evidence, especially using MNK inhibitors in combination with chemotherapy or other targeted therapy, shows promise for future clinical translational studies.

With nearly three decades of research, the knowledge base on mitogen-activated protein kinase (MAPK) interacting kinases or MNKs and related translational efforts toward the development of MNK inhibitors has come a long way. Following the discovery of the eukaryotic translation initiation factor 4E (eIF4E) and its importance in protein translation in the 1980s, researchers identified its activation through the MAPK pathway, specifically by phosphorylation of serine 209 by MNKs.13 Subsequently in the early 2000s, a seminal study by Ueda et al demonstrated that both MNK1 and MNK2 were dispensable for normal cell growth, while this group and others later showed the significance of MNKs in tumorigenesis.4,5 These discoveries suggested that MNKs were potential viable targets in cancer therapy.

Realistically, a single-drug approach is not plausible for most cancer treatments and resistance is a common problem, so drug combinations are utilized. Combinations of mammalian target of rapamycin (mTOR) pathway inhibitors with MNK inhibitors have been previously identified as a potential therapeutic strategy in leukemia.6 The last few years of research surrounding MNK inhibitors had a plethora of studies on dual MNK inhibitors, such as MNK and fms-like tyrosine kinase 3 (FLT3) or MNK and proviral integration site for Moloney murine leukemia virus (PIM) kinase targeting.7,8 Alternatively, more specific MNK inhibitors, such as tomivosertib, are also being explored and have a potential place in combination treatments in efforts to overcome resistance.9 Many groups are still working to develop potent and selective MNK inhibitors with desirable pharmacokinetic properties, while others are focusing on understanding MNK mechanistic elements. There are only three clinical trials that have been initiated utilizing MNK inhibitors in hematological malignancies, but optimized inhibitors and preclinical evidence of combinatorial benefits of MNK inhibitors could promote further clinical research.1012 In this review, we explore the latest updates in pharmacological MNK inhibitor research and how this may apply to clinical studies moving forward.

Leukemias are categorized as acute or chronic and based on the underlying white blood cell type, myeloid or lymphocytic. In general, acute leukemias have a worse prognosis and are more difficult to treat than chronic as evidenced by five-year survival rates for adults.13 The World Health Organization (WHO) 5th edition Haematolymphoid Tumours classification report was just released in 2022 with an aim to clarify myeloid neoplasm diagnosis parameters and categorization criteria to aid physicians in providing more appropriate treatments. Updates in the report for acute myeloid leukemia (AML) incorporate the inclusion of additional mutations, fusions, and rearrangements for defining genetic abnormalities, such as KMT2A, MECOM, and NUP98 rearrangements; additionally, the report explained an AML family restructure to include two categories of defining by genetic abnormalities and defining by differentiation, which allows for diagnosis with genetic abnormalities with less than 20% blasts.14

The standard of care differs amongst the leukemia subtypes. In general, the age and ability of the patient to tolerate intensive treatment stratify the approaches of therapy. Chronic lymphocytic leukemia (CLL), as the least aggressive, if not progressing, does not require treatment immediately and a watch-and-wait approach is used in most cases based on the 2018 guidelines.15,16 However, with a better understanding of the genomic landscape and the discovery of newer targeted therapies with fewer side effects, such as Bruton tyrosine kinase (BTK) inhibitors, there is an increasing number of clinical trials on early intervention strategies for CLL.17 For acute lymphocytic leukemia (ALL), typical treatment starts with chemotherapy regimens of vincristine, dexamethasone, and an anthracycline like doxorubicin, with the possible inclusion of cyclophosphamide and pegaspargase and alternating high-dose methotrexate.18 If it is a Philadelphia (Ph) chromosome positive case, a tyrosine kinase inhibitor may be included, and in T-ALL, nelarabine is sometimes added. In chronic myeloid leukemia (CML), a tyrosine kinase inhibitor such as imatinib or now, more commonly, the second-generation drugs dasatinib, nilotinib, or bosutinib, also BCR::ABL1 inhibitors, are used as first-line therapy in chronic phase CML since the Ph chromosome formation is a hallmark of CML.19 These tyrosine kinase inhibitors have generally been effective for treatment and help prevent progression to the blast phase, but resistance to these inhibitors is possible. For AML, which has the worst prognosis, the traditional treatment for favorable-risk patients is the 7+3 regimen, which consists of two chemotherapy drugs, cytarabine and an anthracycline (daunorubicin, idarubicin), with possible added gemtuzumab ozogamicin.20 However, since this is an intensive treatment regimen, elderly patients or patients with comorbidities may not be able to tolerate it and alternative regimens, such as a combination of a hypomethylating agent with venetoclax, a B-cell lymphoma 2 (BCL2) inhibitor, are often used. Especially with improved profiling techniques, molecular subtypes are even further stratifying treatments. With some of the latest drug developments, specific approvals are sought for certain mutations or patient conditions in AML. For example, olutasidenib, which targets mutant IDH1, showed clinical activity in a phase I/II trial with and without azacytidine and was FDA-approved in December 2022 for relapsed or refractory AML, specifically in patients with an IDH1 mutation.21,22 Similarly, midostaurin, a FLT3 inhibitor, was approved in 2017 for newly diagnosed AML patients with a FLT3 mutation.23 These targeted therapies can provide added benefits to patients that fit the specific criteria. Targeted therapy research, identifying meaningful targets and pursuing the translatability of inhibitors, is prevalent. FDA approvals for targeted drugs in the past five years have included, most recently, olutasidenib (2022), glasdegib targeting the Hedgehog pathway (2018), venetoclax (2018), ivosedinib targeting IDH1 (2018), and midostaurin (2017).24 However, as noted by Estey et al, more approvals are not always better, and the fitness of patients and trial randomization are amongst aspects that should be more rigorously factored in when determining approval and optimal treatment.24 Progress in understanding the genomic landscape, resistance tactics, and beneficial combinational targeting is helping push the field toward more effective leukemia treatments.

MAPK pathways have been of significant interest in cancer research due to the diversity of functional responses mediated by the signaling of this multi-kinase cascade. Further downstream, MNKs are a particularly relevant target in malignancies, due to their vitality in cancer cell signaling but not in normal cell growth and development.4 MNKs are serine/threonine kinases.25 MNK1 and MNK2 both have and isoforms, resulting from alternative splicing where only the isoforms have a MAPK binding domain and a nuclear export sequence.26 Generally, MNK2 has higher basal activity, while MNK1 is more inducible by MAPK signaling.27 Both can be phosphorylated by the MAPKs ERK and p38, but not JNK; MNK2 has preferential binding for ERK while MNK1 has comparable binding to ERK and p38.1 It should be noted that there are negative regulators of MNKs. It has been previously shown that the protein phosphatase 2A dephosphorylates and deactivates MNK1.28 Also, p21 activated kinase 2 (PAK2) phosphorylation was found to disturb the binding of MNK1 to eukaryotic initiation factor 4G (eIF4G) which is the scaffold protein of the eIF4F complex (eIF4G, eIF4E, eIF4A).29 Meanwhile, active MNKs were discovered to bind with mTORC1 and allow for the binding of TELO2 to the complex which instigates mTORC1 downstream substrate phosphorylation.30 To further add to the mTORC1 relation to MNKs, a recent study demonstrated phosphorylation of MNK2 at Ser74 by mTORC1.31 The group showed that this phosphorylation blocked MNK2 binding to eIF4G, thus inhibiting eIF4E phosphorylation by MNK2. This mTORC1-mediated Ser74 phosphorylation is a newly discovered mechanism linking the mTOR and MAPK pathways.

The mRNA 5 cap-binding protein, eIF4E, the limiting factor of the eIF4F complex, is the most well-known target of MNKs and its phosphorylation at serine 209 is used as a readout for MNK activity. Higher phosphorylation of eIF4E correlates with worse prognosis in many cancers and appears to be involved in many oncogenic processes, thereby provoking research targeting the MNK/eIF4E axis.32 A recent study, specifically in AML, found higher nuclear p-eIF4E in primary patient samples was associated with higher tumor burden and worse clinical outcomes.33 Other downstream phosphorylation targets of both MNKs have been identified including Sprouty2, a negative feedback regulator of receptor tyrosine kinases that is phosphorylated at Ser 112 and 121; and the polypyrimidine tract-binding protein-associated splicing factor (PSF), a DNA/RNA binding protein involved in inflammatory cytokine signaling, that is phosphorylated at Ser 8 and 283.3437 MNK1 specific substrates are heterogeneous nuclear ribonucleoprotein A1 (hnRNPA1), an AU-rich element binding protein involved in post-transcriptional regulation, phosphorylated at Ser 192 and Ser 310/311/312, and cytosolic phospholipase A2 (cPLA2), which regulates release of arachidonic acid from glycerophospholipids, phosphorylated at Ser 727 by MNK1.38,39 MNK2 has been shown to phosphorylate the cytoskeleton related protein plectin at Ser 4642.40 Additionally, a recent study demonstrated the translation function of MNK1 specifically in platelets and megakaryocytes where cPLA2 activity was regulated by MNK1, potentially via phosphorylation at serine 505.41 An overview of upstream effectors and downstream targets of MNKs is shown in Figure 1.

Figure 1 Schematic of MNK signaling with the main upstream and downstream proteins and phosphorylation sites identified in the literature. Figure created with BioRender.

Abbreviations: cPLA2, cytosolic phospholipase A2; eIF4E, eukaryotic translation initiation factor 4E; ERK, extracellular signal-regulated kinase; hnRNPA1, heterogeneous nuclear ribonucleoprotein A1; MNK, mitogen-activated protein kinase interacting protein kinase; mTORC1, mammalian target of rapamycin complex 1; PP2A, protein phosphatase 2A; PSF, polypyrimidine tract-binding protein-associated splicing factor; Spry2, sprouty2.

MNKs are part of the Ca2+/calmodulin-dependent kinase group based on structure elements despite not being regulated by either. MNKs have unique structural features that include three short alpha-helices in the catalytic domain, DFD (Asp-Phe-Asp) motifs in the activation loops instead of the usual DFG (Asp-Phe-Gly) motif for other kinases, and an inactive DFD-out conformation with Phe192 in the ATP-binding site which blocks ATP from the catalytic site.42 Asp228 of the DFD motif stabilizes the DFD-out conformation and is unique to MNKs.43 These structural features can be exploited in inhibitor design, especially in targeting the inactive MNK forms. MNK1 and MNK2 have many similarities; the catalytic domains share about 70% of the amino acid framework.44 Kinases most comparable in catalytic domain structure would be ribosomal s6 kinases (RSKs) which are also phosphorylated by ERK, but not p38.44 The unique qualities have led to some hurdles in drug design but that also benefit the development of more specific MNK inhibitors.

There are many MNK inhibitors, developed based on different design approaches, with a range of specificity that have been used in preclinical evaluations. Some MNK inhibitors are already commercially available for laboratory work and are highlighted in Table 1, most of which are type I inhibitors of MNKs. Tomivosertib (eFT508) from eFFECTOR therapeutics is perhaps the most commonly used in recent studies due to its high specificity for MNK1 and MNK2.45 The investigators optimized 4-aminopyrimidines leading to eFT508 which had inhibition of MNK1 and MNK2 and p-eIF4E reduction in the low nanomolar range, high selectivity with only DRAK1 and CLK4 in the off-target kinase profile, and efficacy at reducing tumors in three xenograft mouse models. Optimization of bicyclic fragments with an imidazopyridine derivative exhibiting the greatest MNK inhibition and best pharmacokinetic properties is how ETC-206, now AUM001, was synthesized.46 ETC-206 showed in vitro potency against CML lines and further reduced tumor size in combination with dasatinib in a model of blast crisis CML in mice. Another MNK inhibitor with low nanomolar range inhibition of both MNK1 and MNK2 is SEL201, developed by Selvita and reported in 2017.47 The group discovered this amino-1H-indazol-1,2-dihydropyridin-2-one derivative that they synthesized starting with 5-bromopyridin-2-ol to be an ATP-competitive MNK1/2 inhibitor. Bayer identified an amino-substituted imidazopyridazine by way of high-throughput screening and further optimized this base to design BAY 1142369 which has inhibition of MNK1 and MNK2, with greater activity against MNK1.48 PIM1 was amongst only four other kinases that BAY 1142369 had some inhibitory activity against in the selectivity screening. Another MNK inhibitor, CGP57380, is a pyrazolo-pyrimidine that has shown specificity for MNK1, but the IC50 of MNK1 and p-eIF4E reduction in cells is in the micromolar range.49 Earlier discovered MNK inhibitors were non-specific multi-kinase inhibitors, such as merestinib, which was designed as a type II MET inhibitor but had activity against eleven other kinases that included both MNKs.50 Merestinib mostly targets tyrosine kinases, with MNKs being the only serine/threonine kinases, which they proposed was because of their DFD-out conformation in the inactive form.

Table 1 Commercially Available Synthetically Derived MNK Inhibitors

Despite the existence of commercially available MNK inhibitors, many groups have still been on the hunt for more potent and selective inhibitors with the development of new inhibitors over the past five years. One study screened a fragment library and optimized compounds with a pyridine-(hetero)arylamide core using structure-guided expansion to create selective MNK1/2 inhibitors.51 In general, they found the compounds had greater activity against MNK2 than MNK1 with optimized compound 43 having IC50 values of 89 nM and 200 nM, respectively, and a favorable pharmacokinetic profile. Fragment based design was utilized by another group that designed MNK inhibitors by starting with a benzofuran scaffold and modified it to create 6-hydroxy-4-methoxy-3-methylbenzofuran-7-carboxamide derivatives with top compound 8k having a MNK2 IC50 of 0.27 M and 41% inhibition of MNK1 at 1 M.52 Our group at Northwestern utilized an in silico mutation-based induced-fit docking method and the MNK1 wild-type crystal structure to develop a series of MNK inhibitors that showed activity in the micromolar range against both MNK1 and MNK2 in vitro and reduction of AML cell progenitor growth.42 Another lab designed MNKi-8e, a 5-(2-(phenylamino)pyrimidin-4-yl)thiazole-2(3H)-one derivative, with potent MNK2 inhibition (IC50 = 0.37 M) and antiapoptotic effects in AML cells.53 Jin et al developed 4-((4-fluoro-2-isopropoxyphenyl)amino)-5-methylthieno[2,3-d] pyrimidine derivatives that have MNK inhibitory activity with inhibition of p-eIF4E in cells at 0.1 M and their compound MNK-7g having the most promise.54 These investigators had previously designed a MNK inhibitor, MNK-I1, and were using it as a comparison.

One group has gone through various stages of developing MNK inhibitors over the past ten years. They developed imidazopyridine and imidazopyrazine derivative type I inhibitors using molecular docking to show stable interactions with MNK1 and MNK2 and determined that the importance of MNK inhibitor efficacy was related to the hydrogen bond interaction of the inhibitors and the Lys and Ser residues in the MNK catalytic site.55 Subsequently, they identified type II inhibitors that showed promise through in silico docking simulations with MNK1 and MNK2 IC50 values below 100 nm and generally more potency towards MNK2.56 A majority of MNK inhibitors are type I inhibitors, meaning they are ATP-competitive with the active kinase conformation.57 A recent study took an alternative approach in designing a non-ATP-competitive MNK inhibitor. Bou-Petit et al designed EB1 which has a 4,6-diaryl-1H-pyrazolo[3,4-b]pyridin-3-amine core structure and binds to the inactive MNK1 with an IC50 of 0.69 M (MNK2 IC50 = 9.4 M) preventing transition to its active state and demonstrated reduction of phosphorylation of Ser209 on eIF4E in various cell lines including MV411 AML cells.58 Interestingly, the authors demonstrated that EB1 did not induce upstream activation and phosphorylation of MNK1 leading to enhanced binding to eIF4G as did the type I inhibitors tested in comparison. These results highlight the potential benefits of a type II inhibitor in that it would not lead to paradoxical target protein activation induced by some type I kinase inhibitors which occurs even with catalytic kinase activity inhibition.59 Another group designed non-ATP-competitive inhibitors that were 2H-spiro[cyclohexane-1,3-imidazo[1,5-a]pyridine]-1,5-dione derivatives, using tomivosertib as a comparison; their top prospects had nanomolar inhibition of MNK1 and MNK2 and showed antiproliferative activity in a variety of cancer cell lines.60 Abdelaziz et al also designed a MNK2 inhibitor, an N-phenyl-4-(1H-pyrrol-3-yl)pyrimidin-2-amine derivative, that showed antiproliferative and proapoptotic effects in AML cells.61 Another study designed a MNK1 inhibitor that stabilized the DFD-out conformation of the inactive protein, blocking its activation with a MNK1 IC50 of 21 nM. The reported crystal structure in the Protein Data Bank (5WVD) of the complex of the MNK1 kinase region and the inhibitor could be helpful for future MNK drug design.62 One group analyzed synthesized ferrocene-containing compounds from a known MNK inhibitor and while the added ferrocene made the compounds comparatively more potent at malignant cell inhibition, it negated their MNK1/2 blocking ability.63 Recent work by Halder and Cordeiro led to a multi-targeted in silico screening approach for MNK inhibitors.64 Exploratory model development such as this one could help in better identifying compounds with MNK inhibitory potential.

Not all MNK inhibitors are synthetic in origin; some natural compounds have been discovered to have MNK targeting abilities and some have even been used in cancer clinical trials (Table 2). In general, it is not new to utilize natural compounds in cancer drug discovery, with previous reports indicating around 60% of new drugs having been of natural origin.65 A recent review highlighted over 20 different natural compounds, mostly in preclinical studies, with various antineoplastic effects in leukemia noting the need for therapeutic strategies with less side effects as reasoning for continued analysis of natural sources.66 Most abundantly are studies on various flavonoids which have high contents in fruits, vegetables, herbs, and other plant foods.6769 One study analyzed flavones and flavonols to find kinase inhibitors with AML and ALL cell inhibitory activity and discovered an O-methylated flavonol, a precursor of fisetin, that inhibited MNK2 as well as FLT3, RSK, DYRK2, and JAK2.70 This compound had potent inhibitory effects on AML and ALL cell viability, and induced apoptosis and G0/G1 arrest of AML cells. Myricetin has been shown to inhibit CML cell viability.71 Additionally, a group found that myricetin targeted imatinib-resistant blast phase CML cells by inhibiting activation of eIF4E and demonstrated tumor reduction in a CML-resistant mouse model.72 Chen et al analyzed apigenin, hispidulin, and luteolin, also flavonoids, that all showed nanomolar level inhibition of MNK2, and luteolin also had MNK1 inhibition.73 Additionally, they showed luteolin and hispidulin reduced AML cell growth, increased markers of apoptosis (cleaved caspase 3 and PARP), and arrested cells in sub G1.

Table 2 Commercially Available MNK Inhibitors Found in Natural Sources

Cercosporamide is an antifungal isolated from Cercosporidium henningsii of cassava plants.74 In 2011, it was discovered that it has potent MNK inhibitory activity and has been used in subsequent cancer studies.75 Usnic acid found in lichens has a similar structure to cercosporamide. A study analyzed usnic acid derivatives with a flavanone moiety in leukemia cells and found antiproliferative and antiapoptotic effects while seeing a reduction in phosphorylated eIF4E, but it appeared to be MNK pathway related, not MNK protein specific.76

There have been many studies on the activity of MNK inhibitors in AML models. Our group examined the antileukemic properties of four different commercially available MNK inhibitors, starting with cercosporamide in 2013 which was potent against AML cell lines (U937, MM6) and patient primitive leukemic progenitors while also reducing tumor growth in an MV411 xenograft mouse model.77 In subsequent studies, the lab investigated merestinib, demonstrating inhibition of AML cell progenitor colony formation and additionally saw efficacy in an MM6 mouse model.78 Later, SEL201 was studied, which exhibited antiproliferative and antiapoptotic effects in AML cells.79 Most recently, our group tested tomivosertib in the same established AML cell lines and also saw a reduction in cell viability and colony formation with the highest potency against MV411, MM6 cells, and KG-1 cells.9 Through these four studies, we have found that MNK inhibitors ranging from unselective multi-kinase to very MNK specific all have antineoplastic effects in AML models.

Although not as heavily studied as in AML, MNK inhibitors have been analyzed in CML models. ETC-206 has been identified as a potential MNK inhibitor for use in blast crisis CML with micromolar IC50 values against 25 hematological cell lines including five BCR::ABL1 expressing lines (K562, KCL-22, EM-2, BV-173, JURL-MK1) and enhancement of dasatinib tumor reduction in a blast crisis CML xenograft model.46 Another study analyzed blast crisis CML and leukemia stem cells which they identified to have self-renewal capabilities through -catenin activation, and MNK inhibitors blocked that activation through eIF4E phosphorylation inhibition in vitro and in vivo.80 Resistance of blast crisis CML to tyrosine kinase inhibitors that are typically used for treatment is a major concern so demonstrating a way to target resistant populations is essential.

Recent studies have shown that MNK targeting may be of importance in overcoming resistance in solid tumors both in targeted therapy related instances such as with anlotinib in lung cancer, as well as in chemotherapy related resistance such as with temozolomide in glioblastoma.81,82 Additionally, in gastric cancer, combining tomivosertib with 5-FU or paclitaxel showed benefits in vitro and in vivo, indicating a potential for this MNK inhibitor to sensitize gastric cancer cells to chemotherapy drugs.83 Similarly, in cervical cancer, combination of cercosporamide with chemotherapy drugs, doxorubicin and cisplatin, had increased efficacy in proliferation reduction and apoptosis induction; cercosporamide inhibited chemo-resistant cells and phosphorylation of eIF4E at serine 209 was shown to be induced with chemotherapy treatment.84 In AML, MNK inhibitors have similarly been shown to sensitize cells to the chemotherapy drug cytarabine. We previously reported the activation of the MNK pathway by cytarabine in AML cells and that the combination of the MNK inhibitor, CGP57380, or MNK knockdown with cytarabine enhances the inhibition of leukemic progenitor colony formation.85 Later studies also demonstrated that cytarabine treatment increases MNK activity in AML cells and that there are synergistic effects in AML cells with cytarabine and MNK inhibition (MNKI-8e) or MNK1 and MNK2 knockdown.86 Another group demonstrated that miR-134 was decreased in drug-resistant AML cells (K562/A02, HL-60/ADM) and that MNKs were a target of miR-134; when miR-134 was overexpressed, thereby blocking MNKs, these cells, as well as relapsed AML patient cells, were sensitized to cytarabine.87 Comparatively in CML studies, CML imatinib-resistant cells showed higher sensitivity to myricetin, a plant flavonoid, that reduced phosphorylation of eIF4E at serine 209.72 Similarly, a study showed synergy of CGP57380 with imatinib in inducing apoptosis in Ba/F3 and K562 CML cells.88 Dasatinib, another tyrosine kinase inhibitor used in CML, had synergistic antiproliferative effects in CML cells when combined with niclosamide, which targets the ERK/MNK/eIF4E axis, further demonstrating the importance of the MNK pathway in therapy resistance.89

One of the most common approaches in combinations with MNK inhibitors has been co-targeting with mTOR inhibitors due to the reciprocity of these two pro-survival pathways.6 This has been shown to be a potentially promising approach for a variety of cancers, including in a recent study that demonstrated extended survival using rapamycin, an mTORC1 inhibitor, in combination with tomivosertib or MNK1/2 knockout in an APC KRAS mutated colorectal cancer model.90 In hematological malignancies, a group looking at hematopoietic stem cells found that mTOR deletion lead to increased proliferation and protein synthesis through adaptive signaling by way of MNKs, which may explain the resistance of leukemia cells to mTOR inhibitors and provides reasoning for combination with MNK inhibition; they found mTOR resistant leukemia cells were more sensitive to CGP57380.91 In another study, everolimus, a derivative of rapamycin and also an mTORC1 inhibitor, exhibited synergistic inhibitory effects with CGP57380 in T-ALL cell lines.92 Other work in myeloid leukemia cells showed that phosphorylation of the translational repressor 4EBP1 increases in MNK inhibitor treated cells and there is an enhancement of antiproliferative effects when the MNK1 and MNK2 inhibitors are combined with rapamycin.93 Additionally, a study from our lab showed that SEL201 enhanced the inhibitory effects of rapamycin on AML cells.79 Notably, in the same study it was demonstrated that SEL201 also promoted the effects of 5-azacytidine, a hypomethylating agent approved for use in AML. Cercosporamide was found to enhance rapamycin antileukemic effects in AML cells, while additionally showing synergistic effects with cytarabine both in vitro and in vivo.77 Tomivosertib showed synergistic effects in viability and colony formation reduction with venetoclax, a BCL2 inhibitor approved in CLL and AML in combination with hypomethylating agents or low-dose chemotherapy.9 Combination targets as well as dual MNK inhibitors are highlighted in Table 3.

Table 3 Combination and Dual Targeting with MNK Inhibitors in Preclinical Studies

Typically, in designing inhibitors, it is ideal to demonstrate specificity to one target as it shows minimization of potential off-target effects and mechanistically, higher confidence that an effect is due to the intended inhibited protein. However, in the recent literature surrounding MNKs, several studies are using purposefully designed dual inhibitors. The most common dual inhibition with MNK was with PIMs as the secondary kinase target. PIMs are pro-survival short-lived oncoproteins and are dependent on cap-dependent translation through the eIF4F complex.94 Han et al designed a 4,6-disubstituted pyrido[3,2-d]pyrimidine that had kinase inhibition in the nanomolar range for both MNKs and PIM13.7 One study used extensive in silico analysis to identify three natural compounds from the ZINC database that had high affinity and stable interaction to MNK2 and PIM2 which they suggested as potential therapeutics in AML.95 Usnic acid derivatives were demonstrated to reduce p-eIF4E as well as to target pan PIMs in K562 and HL-60 leukemia cells.76 Although it was not found that MNK was directly inhibited, the reduction to MNK/eIF4E signaling in combination with the PIM inhibition by these compounds further shows the efficacy in targeting these two axes. Virtual screening and docking analysis techniques identified a compound, K783-0308, with high potency and selectivity against MNK2 and FLT3 that was able to inhibit AML cell viability and promote cell cycle arrest.8 Additionally, a BTK/MNK inhibitor, QL-X-138, reduced proliferation in CLL and AML established lines and primary patient cells, noting however the need to improve the pharmacokinetics of the drug for in vivo studies.96 In a study using CML-derived cells, the investigators designed a MNK1/2 and BCR::ABL1 inhibitor with the idea that it would target leukemia stem cells with the MNK inhibition and promote cell death by blocking BCR::ABL1, and they demonstrated both in vitro viability and in vivo tumor reduction efficacy.97 Another group designed 4,6-disubstituted pyrido[3,2-d]pyrimidine derivatives that target both MNK and histone deacetylase (HDAC) which inhibited prostate cancer cell growth and could be tested in leukemia cells since HDAC inhibitors are also being explored individually in leukemia.98,99 The effectiveness of these dual inhibitors could be related to the links of MNKs to overcoming resistance as mentioned in the previous section.

There are a few clinical trials for use of MNK inhibitors in hematological malignancies. Tomivosertib, ETC-1907206, and merestinib are the three MNK inhibitors that were in US clinical trials. Tomivosertib can be found in eight clinical trials with various cancers, with many being in combination studies in advanced cancer state or in combination with immunotherapy.100105 A multicenter hematological malignancy (lymphoma) phase 1 and 2 trial was terminated (NCT02937675), but to the best of our knowledge, no further information has been published at this time.10 ETC-1907206 (ETC-206), now AUM001, was in a phase 1a/1b trial for Ph+ and Ph ALL and Accelerated Phase and Blast Crisis CML in combination with chemotherapy drug dasatinib but was withdrawn (NCT03414450).11 For merestinib, the relapsed or refractory AML phase 1 clinical trial in combination with LY2874455, an FGFR inhibitor, was completed in 2020 with 16 patients (NCT03125239).12 They found merestinib was tolerable and one patient achieved complete remission on the merestinib monotherapy portion of the study.106 Merestinib is a multi-kinase inhibitor and confirmation of its biological activity was based on MET inhibition, so it is unclear as to specific effects of the MNK inhibition.

The current landscape of MNK inhibitor studies is mostly still in the preclinical stage. Only tomivosertib, eFT508, from eFFECTOR Therapeutics has an FDA orphan designation for diffuse large B-cell lymphoma.107 However, there is some promise for the potential future use of MNK inhibitors in combination studies with other agents. Many of the preclinical studies were done in AML models which alludes to a potential for AML clinical trials in the near future. The breadth of mechanistic studies implicates the MNK signaling as a targetable pathway due to its activation in resistant states. This raises the potential of unique clinical-translational approaches targeting MNKs to overcome resistance to chemotherapy and other antileukemia agents.

The research of Dr Platanias is supported by National Institutes of Health grants CA121192, CA77816, NS113425, NS113152 and by grant CX000916 from the Department of Veterans Affairs. Candice Mazewski was supported by NIH/NCI training grant T32 CA070085.

Dr Leonidas C Platanias reports issued patents 10,093,668 and 10,851,082.The authors report no conflicts of interest in this work.

1. Waskiewicz AJ, Flynn A, Proud CG, Cooper JA. Mitogen-activated protein kinases activate the serine/threonine kinases Mnk1 and Mnk2. EMBO J. 1997;16(8):19091920. doi:10.1093/emboj/16.8.1909

2. Bonneau AM, Sonenberg N. Involvement of the 24-kDa cap-binding protein in regulation of protein synthesis in mitosis. J Biol Chem. 1987;262(23):1113411139. doi:10.1016/S0021-9258(18)60935-4

3. Morley SJ, McKendrick L. Involvement of stress-activated protein kinase and p38/RK mitogen-activated protein kinase signaling pathways in the enhanced phosphorylation of initiation factor 4E in NIH 3T3 cells. J Biol Chem. 1997;272(28):1788717893. doi:10.1074/jbc.272.28.17887

4. Ueda T, Watanabe-Fukunaga R, Fukuyama H, Nagata S, Fukunaga R. Mnk2 and Mnk1 are essential for constitutive and inducible phosphorylation of eukaryotic initiation factor 4E but not for cell growth or development. Mol Cell Biol. 2004;24(15):65396549. doi:10.1128/MCB.24.15.6539-6549.2004

5. Ueda T, Sasaki M, Elia AJ, et al. Combined deficiency for MAP kinase-interacting kinase 1 and 2 (Mnk1 and Mnk2) delays tumor development. Proc Natl Acad Sci U S A. 2010;107(32):1398413990. doi:10.1073/pnas.1008136107

6. Kosciuczuk EM, Saleiro D, Platanias LC. Dual targeting of eIF4E by blocking MNK and mTOR pathways in leukemia. Cytokine. 2017;89:116121. doi:10.1016/j.cyto.2016.01.024

7. Han Y, Zhang H, Wang S, et al. Optimization of 4,6-disubstituted pyrido[3,2-d]pyrimidines as dual MNK/PIM inhibitors to inhibit leukemia cell growth. J Med Chem. 2021;64(18):1371913735. doi:10.1021/acs.jmedchem.1c01084

8. Yen SC, Chen LC, Huang HL, et al. Identification of a dual FLT3 and MNK2 inhibitor for acute myeloid leukemia treatment using a structure-based virtual screening approach. Bioorg Chem. 2022;121:105675. doi:10.1016/j.bioorg.2022.105675

9. Suarez M, Blyth GT, Mina AA, et al. Inhibitory effects of Tomivosertib in acute myeloid leukemia. Oncotarget. 2021;12(10):955966. doi:10.18632/oncotarget.27952

10. Therapeutics E. A phase 12 dose-escalation and cohort-expansion study of oral tomivosertib (eFT-508) in subjects with hematological malignancies; 2017. Available from: https://ClinicalTrials.gov/show/NCT02937675. Accessed April 11, 2023.

11. EDDC ASRE, Inc. CI. Evaluation of ETC-1907206 with dasatinib in advanced haematologic malignancies; 2018. Available from: https://ClinicalTrials.gov/show/NCT03414450. Accessed April 11, 2023.

12. Jacqueline Garcia M, Lilly E; Company, Institute D-FC. Combination merestinib and LY2874455 for patients with relapsed or refractory acute myeloid leukemia; 2017. Available from: https://ClinicalTrials.gov/show/NCT03125239. Accessed April 11, 2023.

13. American Cancer Society. Cancer Facts & Figures. American Cancer Society; 2022.

14. Khoury JD, Solary E, Abla O, et al. The 5th edition of the World Health Organization classification of haematolymphoid tumours: myeloid and histiocytic/dendritic neoplasms. Leukemia. 2022;36(7):17031719. doi:10.1038/s41375-022-01613-1

15. Hallek M, Cheson BD, Catovsky D, et al. iwCLL guidelines for diagnosis, indications for treatment, response assessment, and supportive management of CLL. Blood. 2018;131(25):27452760. doi:10.1182/blood-2017-09-806398

16. Hampel PJ, Parikh SA. Chronic lymphocytic leukemia treatment algorithm 2022. Blood Cancer J. 2022;12(11):161. doi:10.1038/s41408-022-00756-9

17. Kay NE, Parikh SA, Parikh SA. Early intervention in asymptomatic chronic lymphocytic leukemia. Clin Adv Hematol Oncol. 2021;19(2):92103.

18. American Cancer Society. Treating Acute Lymphocytic Leukemia (ALL). American Cancer Society; 2021.

19. Deininger MW, Shah NP, Altman JK, et al. Chronic myeloid leukemia, version 2.2021, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 2020;18(10):13851415. doi:10.6004/jnccn.2020.0047

20. Tallman MS, Wang ES, Altman JK, et al. Acute myeloid leukemia, version 3.2019, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 2019;17(6):721749. doi:10.6004/jnccn.2019.0028

21. Watts JM, Baer MR, Yang J, et al. Olutasidenib alone or with azacitidine in IDH1-mutated acute myeloid leukaemia and myelodysplastic syndrome: phase 1 results of a phase 1/2 trial. Lancet Haematol. 2022;10:e46e58.

22. Administration FaD. FDA Approves Olutasidenib for Relapsed or Refractory Acute Myeloid Leukemia with a Susceptible IDH1 Mutation. Administration FaD; 2023:ed2022.

23. Levis M. Midostaurin approved for FLT3-mutated AML. Blood. 2017;129(26):34033406. doi:10.1182/blood-2017-05-782292

24. Estey E, Karp JE, Emadi A, Othus M, Gale RP. Recent drug approvals for newly diagnosed acute myeloid leukemia: gifts or a Trojan horse? Leukemia. 2020;34(3):671681. doi:10.1038/s41375-019-0704-5

25. Fukunaga R, Hunter T. MNK1, a new MAP kinase-activated protein kinase, isolated by a novel expression screening method for identifying protein kinase substrates. EMBO J. 1997;16(8):19211933. doi:10.1093/emboj/16.8.1921

26. Buxade M, Parra-Palau JL, Proud CG. The Mnks: MAP kinase-interacting kinases (MAP kinase signal-integrating kinases). Front Biosci. 2008;13(14):53595374. doi:10.2741/3086

27. Scheper GC, Morrice NA, Kleijn M, Proud CG. The mitogen-activated protein kinase signal-integrating kinase Mnk2 is a eukaryotic initiation factor 4E kinase with high levels of basal activity in mammalian cells. Mol Cell Biol. 2001;21(3):743754. doi:10.1128/MCB.21.3.743-754.2001

28. Li Y, Yue P, Deng X, et al. Protein phosphatase 2A negatively regulates eukaryotic initiation factor 4E phosphorylation and eIF4F assembly through direct dephosphorylation of Mnk and eIF4E. Neoplasia. 2010;12(10):848855. doi:10.1593/neo.10704

29. Orton KC, Ling J, Waskiewicz AJ, et al. Phosphorylation of Mnk1 by caspase-activated Pak2/gamma-PAK inhibits phosphorylation and interaction of eIF4G with Mnk. J Biol Chem. 2004;279(37):3864938657. doi:10.1074/jbc.M407337200

30. Brown MC, Gromeier M. MNK controls mTORC1: substrate association through regulation of TELO2 binding with mTORC1. Cell Rep. 2017;18(6):14441457. doi:10.1016/j.celrep.2017.01.023

31. Xie J, Shen K, Jones AT, et al. Reciprocal signaling between mTORC1 and MNK2 controls cell growth and oncogenesis. Cell Mol Life Sci. 2021;78:249270. doi:10.1007/s00018-020-03491-1

32. Yang X, Zhong W, Cao R. Phosphorylation of the mRNA cap-binding protein eIF4E and cancer. Cell Signal. 2020;73:109689. doi:10.1016/j.cellsig.2020.109689

33. Zhou H, Jia X, Yang FAN. Elevated nuclear phospho-eIF4E body levels are associated with tumor progression and poor prognosis for acute myeloid leukemia. Biocell. 2021;45(3):711722. doi:10.32604/biocell.2021.014193

34. Joshi S, Platanias LC. Mnk kinase pathway: cellular functions and biological outcomes. World J Biol Chem. 2014;5(3):321333. doi:10.4331/wjbc.v5.i3.321

35. DaSilva J, Xu L, Kim HJ, Miller WT, Bar-Sagi D. Regulation of sprouty stability by Mnk1-dependent phosphorylation. Mol Cell Biol. 2006;26(5):18981907. doi:10.1128/MCB.26.5.1898-1907.2006

36. Edwin F, Anderson K, Patel TB. HECT domain-containing E3 ubiquitin ligase Nedd4 interacts with and ubiquitinates Sprouty2. J Biol Chem. 2010;285(1):255264. doi:10.1074/jbc.M109.030882

37. Buxade M, Morrice N, Krebs DL, Proud CG. The PSF.p54nrb complex is a novel Mnk substrate that binds the mRNA for tumor necrosis factor alpha. J Biol Chem. 2008;283(1):5765. doi:10.1074/jbc.M705286200

38. Buxade M, Parra JL, Rousseau S, et al. The Mnks are novel components in the control of TNF alpha biosynthesis and phosphorylate and regulate hnRNP A1. Immunity. 2005;23(2):177189. doi:10.1016/j.immuni.2005.06.009

39. Hefner Y, Borsch-Haubold AG, Murakami M, et al. Serine 727 phosphorylation and activation of cytosolic phospholipase A2 by MNK1-related protein kinases. J Biol Chem. 2000;275(48):3754237551. doi:10.1074/jbc.M003395200

40. Bouameur JE, Schneider Y, Begre N, et al. Phosphorylation of serine 4642 in the C-terminus of plectin by MNK2 and PKA modulates its interaction with intermediate filaments. J Cell Sci. 2013;126(Pt 18):41954207. doi:10.1242/jcs.127779

41. Manne BK, Campbell RA, Bhatlekar S, et al. MAPK-interacting kinase 1 regulates platelet production, activation, and thrombosis. Blood. 2022;140(23):24772489. doi:10.1182/blood.2022015568

42. Mishra RK, Clutter MR, Blyth GT, et al. Discovery of novel Mnk inhibitors using mutation-based induced-fit virtual high-throughput screening. Chem Biol Drug Des. 2019;94(4):18131823. doi:10.1111/cbdd.13585

43. Jauch R, Jakel S, Netter C, et al. Crystal structures of the Mnk2 kinase domain reveal an inhibitory conformation and a zinc binding site. Structure. 2005;13(10):15591568. doi:10.1016/j.str.2005.07.013

44. Cargnello M, Roux PP. Activation and function of the MAPKs and their substrates, the MAPK-activated protein kinases. Microbiol Mol Biol Rev. 2011;75(1):5083. doi:10.1128/MMBR.00031-10

45. Reich SH, Sprengeler PA, Chiang GG, et al. Structure-based design of pyridone-aminal eFT508 targeting dysregulated translation by selective mitogen-activated protein kinase interacting kinases 1 and 2 (MNK1/2) Inhibition. J Med Chem. 2018;61(8):35163540. doi:10.1021/acs.jmedchem.7b01795

46. Yang H, Chennamaneni LR, Ho MWT, et al. Optimization of selective mitogen-activated protein kinase interacting kinases 1 and 2 inhibitors for the treatment of blast crisis leukemia. J Med Chem. 2018;61(10):43484369. doi:10.1021/acs.jmedchem.7b01714

47. Zhan Y, Guo J, Yang W, et al. MNK1/2 inhibition limits oncogenicity and metastasis of KIT-mutant melanoma. J Clin Invest. 2017;127(11):41794192. doi:10.1172/JCI91258

48. Santag S, Siegel F, Wengner AM, et al. BAY 1143269, a novel MNK1 inhibitor, targets oncogenic protein expression and shows potent anti-tumor activity. Cancer Lett. 2017;390:2129. doi:10.1016/j.canlet.2016.12.029

49. Knauf U, Tschopp C, Gram H. Negative regulation of protein translation by mitogen-activated protein kinase-interacting kinases 1 and 2. Mol Cell Biol. 2001;21(16):55005511. doi:10.1128/MCB.21.16.5500-5511.2001

50. Yan SB, Peek VL, Ajamie R, et al. LY2801653 is an orally bioavailable multi-kinase inhibitor with potent activity against MET, MST1R, and other oncoproteins, and displays anti-tumor activities in mouse xenograft models. Invest New Drugs. 2013;31(4):833844. doi:10.1007/s10637-012-9912-9

51. Kwiatkowski J, Liu B, Pang S, et al. Stepwise evolution of fragment hits against MAPK interacting kinases 1 and 2. J Med Chem. 2020;63(2):621637. doi:10.1021/acs.jmedchem.9b01582

52. Wang S, Li B, Liu B, et al. Design and synthesis of novel 6-hydroxy-4-methoxy-3-methylbenzofuran-7-carboxamide derivatives as potent Mnks inhibitors by fragment-based drug design. Bioorg Med Chem. 2018;26(16):46024614. doi:10.1016/j.bmc.2018.05.004

53. Diab S, Teo T, Kumarasiri M, et al. Discovery of 5-(2-(Phenylamino)pyrimidin-4-yl)thiazol-2(3H)-one derivatives as potent Mnk2 inhibitors: synthesis, SAR analysis and biological evaluation. ChemMedChem. 2014;9(5):962972. doi:10.1002/cmdc.201300552

54. Jin X, Merrett J, Tong S, et al. Design, synthesis and activity of Mnk1 and Mnk2 selective inhibitors containing thieno[2,3-d]pyrimidine scaffold. Eur J Med Chem. 2019;162:735751. doi:10.1016/j.ejmech.2018.10.070

55. Kannan S, Poulsen A, Yang HY, et al. Probing the binding mechanism of Mnk inhibitors by docking and molecular dynamics simulations. Biochemistry. 2015;54(1):3246. doi:10.1021/bi501261j

56. Kannan S, Pradhan MR, Cherian J, et al. Small molecules targeting the inactive form of the Mnk1/2 kinases. ACS Omega. 2017;2(11):78817891. doi:10.1021/acsomega.7b01403

57. Xu W, Kannan S, Verma CS, Nacro K. Update on the development of MNK inhibitors as therapeutic agents. J Med Chem. 2022;65(2):9831007. doi:10.1021/acs.jmedchem.1c00368

58. Bou-Petit E, Hummer S, Alarcon H, et al. Overcoming paradoxical kinase priming by a novel MNK1 inhibitor. J Med Chem. 2022;65(8):60706087. doi:10.1021/acs.jmedchem.1c01941

59. Hantschel O. Unexpected off-targets and paradoxical pathway activation by kinase inhibitors. ACS Chem Biol. 2015;10(1):234245. doi:10.1021/cb500886n

60. Abdelaziz AM, Basnet SKC, Islam S, et al. Synthesis and evaluation of 2H-spiro[cyclohexane-1,3-imidazo[1,5-a]pyridine]-1,5-dione derivatives as Mnk inhibitors. Bioorg Med Chem Lett. 2019;29(18):26502654. doi:10.1016/j.bmcl.2019.07.043

61. Abdelaziz AM, Diab S, Islam S, et al. Discovery of N-phenyl-4-(1H-pyrrol-3-yl)pyrimidin-2-amine derivatives as potent Mnk2 inhibitors: design, synthesis, SAR analysis, and evaluation of in vitro anti-leukaemic activity. Med Chem. 2019;15(6):602623. doi:10.2174/1573406415666181219111511

62. Matsui Y, Yasumatsu I, Yoshida KI, et al. A novel inhibitor stabilizes the inactive conformation of MAPK-interacting kinase 1. Acta Crystallogr Struct Biol Commun. 2018;74(Pt 3):156160. doi:10.1107/S2053230X18002108

63. Sansook S, Lineham E, Hassell-Hart S, et al. Probing the anticancer action of novel ferrocene analogues of MNK inhibitors. Molecules. 2018;23(9):2126. doi:10.3390/molecules23092126

64. Halder AK, Cordeiro M. Multi-target in silico prediction of inhibitors for mitogen-activated protein kinase-interacting kinases. Biomolecules. 2021;11(11):1670. doi:10.3390/biom11111670

65. Newman DJ, Cragg GM, Snader KM. Natural products as sources of new drugs over the period 19812002. J Nat Prod. 2003;66:10221037. doi:10.1021/np030096l

66. Cotoraci C, Ciceu A, Sasu A, Miutescu E, Hermenean A. The anti-leukemic activity of natural compounds. Molecules. 2021;26(9):2709. doi:10.3390/molecules26092709

67. Kimira M, Arai Y, Shimoi K, Watanabe S. Japanese intake of flavonoids and isoflavonoids from foods. J Epidemiol. 1998;8(3):168175. doi:10.2188/jea.8.168

68. Bower AM, Real Hernandez LM, Berhow MA, de Mejia EG. Bioactive compounds from culinary herbs inhibit a molecular target for type 2 diabetes management, dipeptidyl peptidase IV. J Agric Food Chem. 2014;62(26):61476158. doi:10.1021/jf500639f

69. Shankar E, Goel A, Gupta K, Gupta S. Plant flavone apigenin: an emerging anticancer agent. Curr Pharmacol Rep. 2017;3(6):423446. doi:10.1007/s40495-017-0113-2

70. Yen SC, Wu YW, Huang CC, et al. O-methylated flavonol as a multi-kinase inhibitor of leukemogenic kinases exhibits a potential treatment for acute myeloid leukemia. Phytomedicine. 2022;100:154061. doi:10.1016/j.phymed.2022.154061

71. Pan H, Hu Q, Wang J, et al. Myricetin is a novel inhibitor of human inosine 5-monophosphate dehydrogenase with anti-leukemia activity. Biochem Biophys Res Commun. 2016;477(4):915922. doi:10.1016/j.bbrc.2016.06.158

72. Cai F, Li B, Li J, Ding Y, Xu D, Huang F. Myricetin is effective and selective in inhibiting imatinib-resistant chronic myeloid leukemia stem and differentiated cells through targeting eIF4E. Anticancer Drugs. 2022;34:620626. doi:10.1097/CAD.0000000000001421

See the rest here:
MNK proteins as therapeutic targets in leukemia | OTT - Dove Medical Press

Novel chemokine related LncRNA signature correlates with the … – BMC Gastroenterology

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394424.

Article PubMed Google Scholar

Arnold M, Soerjomataram I, Ferlay J, Forman D. Global incidence of oesophageal cancer by histological subtype in 2012. Gut. 2015;64(3):3817.

Article PubMed Google Scholar

Hersznyi L, Tulassay Z. Epidemiology of gastrointestinal and liver tumors. Eur Rev Med Pharmacol Sci. 2010;14(4):24958.

PubMed Google Scholar

Wang WL, Chang WL, Yang HB, Wang YC, Chang IW, Lee CT, Chang CY, Lin JT, Sheu BS. Low disabled-2 expression promotes tumor progression and determines poor survival and high recurrence of esophageal squamous cell carcinoma. Oncotarget. 2016;7(44):7116981.

Article PubMed PubMed Central Google Scholar

Aquino JL, Said MM, Pereira DA, Cecchino GN, Leandro-Merhi VA. Complications of the rescue esophagectomy in advanced esophageal cancer. Arq Bras Cir Dig. 2013;26(3):1738 ABCD = Brazilian archives of digestive surgery.

Article PubMed Google Scholar

Zhang J, Ling X, Fang C, Ma J. Identification and validation of an eight-lncRNA signature that predicts prognosis in patients with esophageal squamous cell carcinoma. Cell Mol Biol Lett. 2022;27(1):39.

Article CAS PubMed PubMed Central Google Scholar

Zheng ZJ, Li YS, Zhu JD, Zou HY, Fang WK, Cui YY, Xie JJ. Construction of the Six-lncRNA prognosis signature as a novel biomarker in esophageal squamous cell carcinoma. Front Genet. 2022;13: 839589.

Article CAS PubMed PubMed Central Google Scholar

Chi Y, Wang D, Wang J, Yu W, Yang J. Long Non-Coding RNA in the pathogenesis of cancers. Cells. 2019;8(9):1015.

Article CAS PubMed PubMed Central Google Scholar

Fang P, Chen H, Ma Z, Han C, Yin W, Wang S, Zhu H, Xia W, Wang J, Xu L, et al. lncRNA LINC00525 suppresses p21 expression via mRNA decay and triplex-mediated changes in chromatin structure in lung adenocarcinoma. Cancer Commun (London, England). 2021;41(7):596614.

Article Google Scholar

Sheykhhasan M, Ahmadyousefi Y, Seyedebrahimi R, Tanzadehpanah H, Manoochehri H, Dama P, Hosseini NF, Akbari M, Eslami Farsani M. DLX6-AS1: a putative lncRNA candidate in multiple human cancers. Expert Rev Mol Med. 2021;23: e17.

Article CAS PubMed Google Scholar

Bhan A, Soleimani M, Mandal SS. Long Noncoding RNA and Cancer: A New Paradigm. Can Res. 2017;77(15):396581.

Article CAS Google Scholar

Zhu J, Zhao Y, Wu G, Zhang X, Chen Q, Yang B, Guo X, Ji S, Gu K. Ferroptosis-related lncRNA signature correlates with the prognosis, tumor microenvironment, and therapeutic sensitivity of esophageal squamous cell carcinoma. Oxid Med Cell Longev. 2022;2022:7465880.

Article PubMed PubMed Central Google Scholar

Zhao F, Li Y, Dong Z, Zhang D, Guo P, Li Z, Li S. Identification of a risk signature based on lactic acid metabolism-related lncRNAs in patients with esophageal squamous cell carcinoma. Front Cell Dev Biol. 2022;10:845293.

Article PubMed PubMed Central Google Scholar

Zhao F, Dong Z, Li Y, Liu S, Guo P, Zhang D, Li S. Comprehensive analysis of molecular clusters and prognostic signature based on m7G-related lncRNAs in esophageal squamous cell carcinoma. Front Oncol. 2022;12: 893186.

Article PubMed PubMed Central Google Scholar

Shi X, Liu X, Pan S, Ke Y, Li Y, Guo W, Wang Y, Ruan Q, Zhang X, Ma H. A novel autophagy-related long non-coding RNA signature to predict prognosis and therapeutic response in esophageal squamous cell carcinoma. Int J Gen Med. 2021;14:832539.

Article CAS PubMed PubMed Central Google Scholar

Zhu T, Ma Z, Wang H, Wei D, Wang B, Zhang C, Fu L, Li Z, Yu G. Immune-related long non-coding RNA signature and clinical nomogram to evaluate survival of patients suffering esophageal squamous cell carcinoma. Front Cell Dev Biol. 2021;9:641960.

Article PubMed PubMed Central Google Scholar

Mantovani A, Savino B, Locati M, Zammataro L, Allavena P, Bonecchi R. The chemokine system in cancer biology and therapy. Cytokine Growth Factor Rev. 2010;21(1):2739.

Article CAS PubMed Google Scholar

Nagarsheth N, Wicha MS, Zou W. Chemokines in the cancer microenvironment and their relevance in cancer immunotherapy. Nat Rev Immunol. 2017;17(9):55972.

Article CAS PubMed PubMed Central Google Scholar

Dangaj D, Bruand M, Grimm AJ, Ronet C, Barras D, Duttagupta PA, Lanitis E, Duraiswamy J, Tanyi JL, Benencia F, et al. Cooperation between constitutive and inducible chemokines enables T cell engraftment and immune attack in solid tumors. Cancer Cell. 2019;35(6):885-900.e810.

Article CAS PubMed PubMed Central Google Scholar

Lim SJ. CCL24 signaling in the tumor microenvironment. Adv Exp Med Biol. 2021;1302:918.

Article PubMed Google Scholar

Harrow J, Frankish A, Gonzalez JM, Tapanari E, Diekhans M, Kokocinski F, Aken BL, Barrell D, Zadissa A, Searle S, et al. GENCODE: the reference human genome annotation for the ENCODE project. Genome Res. 2012;22(9):176074.

Article CAS PubMed PubMed Central Google Scholar

White NM, Cabanski CR, Silva-Fisher JM, Dang HX, Govindan R, Maher CA. Transcriptome sequencing reveals altered long intergenic non-coding RNAs in lung cancer. Genome Biol. 2014;15(8):429.

Article PubMed PubMed Central Google Scholar

Guo JC, Wu Y, Chen Y, Pan F, Wu ZY, Zhang JS, Wu JY, Xu XE, Zhao JM, Li EM, et al. Protein-coding genes combined with long noncoding RNA as a novel transcriptome molecular staging model to predict the survival of patients with esophageal squamous cell carcinoma. Cancer Commun (London, England). 2018;38(1):4.

Article Google Scholar

Zlotnik A, Yoshie O. The chemokine superfamily revisited. Immunity. 2012;36(5):70516.

Article CAS PubMed PubMed Central Google Scholar

Griffith JW, Sokol CL, Luster AD. Chemokines and chemokine receptors: positioning cells for host defense and immunity. Annu Rev Immunol. 2014;32:659702.

Article CAS PubMed Google Scholar

Sokol CL, Luster AD. The chemokine system in innate immunity. Cold Spring Harb Perspect Biol. 2015;7(5):a016303.

Article PubMed PubMed Central Google Scholar

Tiberio L, Del Prete A, Schioppa T, Sozio F, Bosisio D, Sozzani S. Chemokine and chemotactic signals in dendritic cell migration. Cell Mol Immunol. 2018;15(4):34652.

Article CAS PubMed PubMed Central Google Scholar

Wang X, Zhao Y, Strohmer DF, Yang W, Xia Z, Yu C. The prognostic value of MicroRNAs associated with fatty acid metabolism in head and neck squamous cell carcinoma. Front Genet. 2022;13: 983672.

Article CAS PubMed PubMed Central Google Scholar

Kanehisa M, Goto S. KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 2000;28(1):2730.

Article CAS PubMed PubMed Central Google Scholar

Kanehisa M. Toward understanding the origin and evolution of cellular organisms. Protein Sci. 2019;28(11):194751.

Article CAS PubMed PubMed Central Google Scholar

Kanehisa M, Furumichi M, Sato Y, Ishiguro-Watanabe M, Tanabe M. KEGG: integrating viruses and cellular organisms. Nucleic Acids Res. 2021;49(D1):D545-d551.

Article CAS PubMed Google Scholar

Chi H, Xie X, Yan Y, Peng G, Strohmer DF, Lai G, Zhao S, Xia Z, Tian G. Natural killer cell-related prognosis signature characterizes immune landscape and predicts prognosis of HNSCC. Front Immunol. 2022;13:1018685.

Article CAS PubMed PubMed Central Google Scholar

Chi H, Jiang P, Xu K, Zhao Y, Song B, Peng G, He B, Liu X, Xia Z, Tian G. A novel anoikis-related gene signature predicts prognosis in patients with head and neck squamous cell carcinoma and reveals immune infiltration. Front Genet. 2022;13: 984273.

Article CAS PubMed PubMed Central Google Scholar

Korbecki J, Kupnicka P, Chlubek M, Gorcy J, Gutowska I, Baranowska-Bosiacka I. CXCR2 receptor: regulation of expression, signal transduction, and involvement in Cancer. Int J Mol Sci. 2022;23(4):2168.

Article CAS PubMed PubMed Central Google Scholar

Bill CA, Allen CM, Vines CM. C-C Chemokine receptor 7 in cancer. Cells. 2022;11(4):656.

Article CAS PubMed PubMed Central Google Scholar

Gong X, Chi H, Strohmer DF, Teichmann AT, Xia Z, Wang Q. Exosomes: a potential tool for immunotherapy of ovarian cancer. Front Immunol. 2023;13:1089410.

Article PubMed PubMed Central Google Scholar

Yura M, Fukuda K, Matsuda S, Irino T, Nakamura R, Kawakubo H, Takeuchi H, Kitagawa Y. Effects of let-7a microRNA and C-C chemokine receptor type 7 expression on cellular function and prognosis in esophageal squamous cell carcinoma. BMC Cancer. 2022;22(1):1064.

Article CAS PubMed PubMed Central Google Scholar

Guo J, Tong CY, Shi JG, Li XJ. C-X-C motif chemokine ligand 12 (CXCL12)/C-X-C motif chemokine receptor 7(CXCR7) regulates epithelial-mesenchymal transition process and promotes the metastasis of esophageal cancer by activating signal transducer and activator of transcription 3 (STAT3) pathway. Bioengineered. 2022;13(3):742538.

Article CAS PubMed PubMed Central Google Scholar

Zhong YB, Shan AJ, Lv W, Wang J, Xu JZ. Long non-coding RNA LINC00675 inhibits tumorigenesis and EMT via repressing Wnt/-catenin signaling in esophageal squamous cell carcinoma. Eur Rev Med Pharmacol Sci. 2018;22(23):828897.

PubMed Google Scholar

Zhu Z, Wang H, Pang Y, Hu H, Zhang H, Wang W. Exosomal long non-coding RNA UCA1 functions as growth inhibitor in esophageal cancer. Aging. 2020;12(20):8581.

Article Google Scholar

Wang X, Gao Z, Liao J, Shang M, Li X, Yin L, Pu Y, Liu R. lncRNA UCA1 inhibits esophageal squamous-cell carcinoma growth by regulating the Wnt signaling pathway. J Toxicol Environ Health Part A. 2016;79(910):40718.

Article CAS Google Scholar

Wang X, Sun M, Gao Z, Yin L, Pu Y, Zhu Y, Wang X, Liu R. N-nitrosamines-mediated downregulation of lncRNA-UCA1 induces carcinogenesis of esophageal squamous by regulating the alternative splicing of FGFR2. Sci Total Environ. 2023;855: 158918.

Article CAS PubMed Google Scholar

Gautam SK, Basu S, Aithal A, Dwivedi NV, Gulati M, Jain M. Regulation of pancreatic cancer therapy resistance by chemokines. Semin Cancer Biol. 2022;86(Pt 2):6980.

Article CAS PubMed Google Scholar

Bhat AA, Nisar S, Maacha S, Carneiro-Lobo TC, Akhtar S, Siveen KS, Wani NA, Rizwan A, Bagga P, Singh M, et al. Cytokine-chemokine network driven metastasis in esophageal cancer; promising avenue for targeted therapy. Mol Cancer. 2021;20(1):2.

Article PubMed PubMed Central Google Scholar

Fujikawa M, Koma YI, Hosono M, Urakawa N, Tanigawa K, Shimizu M, Kodama T, Sakamoto H, Nishio M, Shigeoka M, et al. Chemokine (C-C Motif) Ligand 1 derived from tumor-associated macrophages contributes to esophageal squamous cell carcinoma progression via CCR8-mediated Akt/Proline-rich Akt substrate of 40 kDa/Mammalian target of rapamycin pathway. Am J Pathol. 2021;191(4):686703.

Article CAS PubMed Google Scholar

Zhao Y, Wei K, Chi H, Xia Z, Li X. IL-7: a promising adjuvant ensuring effective T cell responses and memory in combination with cancer vaccines? Front Immunol. 2022;13:1022808.

Article CAS PubMed PubMed Central Google Scholar

Ugel S, Can S, De Sanctis F, Bronte V. Monocytes in the tumor microenvironment. Annu Rev Pathol. 2021;16:93122.

Article CAS PubMed Google Scholar

Cassetta L, Pollard JW. Targeting macrophages: therapeutic approaches in cancer. Nat Rev Drug Discovery. 2018;17(12):887904.

Article CAS PubMed Google Scholar

Wu Z, Zhang X, Chen D, Li Z, Wu X, Wang J, Deng Y. N6-Methyladenosine-related lncRNAs are potential remodeling indicators in the tumor microenvironment and prognostic markers in osteosarcoma. Front Immunol. 2021;12: 806189.

Article CAS PubMed Google Scholar

Zhou R, Liang J, Tian H, Chen Q, Yang C, Liu C. Development of a Ferroptosis-related lncRNA signature to predict the prognosis and immune landscape of bladder cancer. Dis Markers. 2021;2021:1031906.

Article PubMed PubMed Central Google Scholar

Read more:
Novel chemokine related LncRNA signature correlates with the ... - BMC Gastroenterology

Long- and very long-chain ceramides are predictors of acute kidney … – Cardiovascular Diabetology

Marenzi G, Cosentino N, Bartorelli AL. Acute kidney injury in patients with acute coronary syndromes. Heart. 2015;101(22):177885.

Article CAS PubMed Google Scholar

Marenzi G, Cabiati A, Bertoli SV, Assanelli E, Marana I, De Metrio M, Rubino M, Moltrasio M, Grazi M, Campodonico J, et al. Incidence and relevance of acute kidney injury in patients hospitalized with acute coronary syndromes. Am J Cardiol. 2013;111(6):81622.

Article PubMed Google Scholar

Goldberg A, Hammerman H, Petcherski S, Zdorovyak A, Yalonetsky S, Kapeliovich M, Agmon Y, Markiewicz W, Aronson D. Inhospital and 1-year mortality of patients who develop worsening renal function following acute ST-elevation myocardial infarction. Am Heart J. 2005;150(2):3307.

Article PubMed Google Scholar

Kaltsas E, Chalikias G, Tziakas D. The incidence and the prognostic impact of acute kidney injury in acute myocardial infarction patients: current preventive strategies. Cardiovasc Drugs Ther. 2018;32(1):8198.

Article PubMed Google Scholar

Marenzi G, Assanelli E, Campodonico J, De Metrio M, Lauri G, Marana I, Moltrasio M, Rubino M, Veglia F, Montorsi P, et al. Acute kidney injury in ST-segment elevation acute myocardial infarction complicated by cardiogenic shock at admission. Crit Care Med. 2010;38(2):43844.

Article PubMed Google Scholar

Gault CR, Obeid LM, Hannun YA. An overview of sphingolipid metabolism: from synthesis to breakdown. Adv Exp Med Biol. 2010;688:123.

Article CAS PubMed PubMed Central Google Scholar

Anroedh S, Hilvo M, Akkerhuis KM, Kauhanen D, Koistinen K, Oemrawsingh R, Serruys P, van Geuns RJ, Boersma E, Laaksonen R, et al. Plasma concentrations of molecular lipid species predict long-term clinical outcome in coronary artery disease patients. J Lipid Res. 2018;59(9):172937.

Article CAS PubMed PubMed Central Google Scholar

Cheng JM, Suoniemi M, Kardys I, Vihervaara T, de Boer SP, Akkerhuis KM, Sysi-Aho M, Ekroos K, Garcia-Garcia HM, Oemrawsingh RM, et al. Plasma concentrations of molecular lipid species in relation to coronary plaque characteristics and cardiovascular outcome: results of the ATHEROREMO-IVUS study. Atherosclerosis. 2015;243(2):5606.

Article CAS PubMed Google Scholar

Laaksonen R, Ekroos K, Sysi-Aho M, Hilvo M, Vihervaara T, Kauhanen D, Suoniemi M, Hurme R, Mrz W, Scharnagl H, et al. Plasma ceramides predict cardiovascular death in patients with stable coronary artery disease and acute coronary syndromes beyond LDL-cholesterol. Eur Heart J. 2016;37(25):196776.

Article CAS PubMed PubMed Central Google Scholar

Tu C, Xie L, Wang Z, Zhang L, Wu H, Ni W, Li C, Li L, Zeng Y. Association between ceramides and coronary artery stenosis in patients with coronary artery disease. Lipids Health Dis. 2020;19(1):151.

Article CAS PubMed PubMed Central Google Scholar

Peterson LR, Xanthakis V, Duncan MS, Gross S, Friedrich N, Volzke H, Felix SB, Jiang H, Sidhu R, Nauck M, et al. Ceramide remodeling and risk of cardiovascular events and mortality. J Am Heart Assoc. 2018. https://doi.org/10.1161/JAHA.117.007931.

Article PubMed PubMed Central Google Scholar

Tarasov K, Ekroos K, Suoniemi M, Kauhanen D, Sylvnne T, Hurme R, Gouni-Berthold I, Berthold HK, Kleber ME, Laaksonen R, et al. Molecular lipids identify cardiovascular risk and are efficiently lowered by simvastatin and PCSK9 deficiency. J Clin Endocrinol Metab. 2014;99(1):E45-52.

Article PubMed Google Scholar

Nwabuo CC, Duncan M, Xanthakis V, Peterson LR, Mitchell GF, McManus D, Cheng S, Vasan RS. Association of circulating ceramides with cardiac structure and function in the community: the framingham heart study. J Am Heart Assoc. 2019;8(19):e013050.

Article PubMed PubMed Central Google Scholar

Li PL, Zhang Y. Cross talk between ceramide and redox signaling: implications for endothelial dysfunction and renal disease. Handb Exp Pharmacol. 2013;216:17197.

Article CAS Google Scholar

Basnakian AG, Ueda N, Hong X, Galitovsky VE, Yin X, Shah SV. Ceramide synthase is essential for endonuclease-mediated death of renal tubular epithelial cells induced by hypoxia-reoxygenation. Am J Physiol Renal Physiol. 2005;288(2):F308-314.

Article CAS PubMed Google Scholar

Ueda N. Ceramide-induced apoptosis in renal tubular cells: a role of mitochondria and sphingosine-1-phoshate. Int J Mol Sci. 2015;16(3):5076124.

Article CAS PubMed PubMed Central Google Scholar

Ostermann M, Liu K. Pathophysiology of AKI. Best Pract Res Clin Anaesthesiol. 2017;31(3):30514.

Article PubMed Google Scholar

Raichur S, Brunner B, Bielohuby M, Hansen G, Pfenninger A, Wang B, Bruning JC, Larsen PJ, Tennagels N. The role of C16:0 ceramide in the development of obesity and type 2 diabetes: CerS6 inhibition as a novel therapeutic approach. Mol Metab. 2019;21:3650.

Article CAS PubMed PubMed Central Google Scholar

Turpin SM, Nicholls HT, Willmes DM, Mourier A, Brodesser S, Wunderlich CM, Mauer J, Xu E, Hammerschmidt P, Bronneke HS, et al. Obesity-induced CerS6-dependent C16:0 ceramide production promotes weight gain and glucose intolerance. Cell Metab. 2014;20(4):67886.

Article CAS PubMed Google Scholar

Fekry B, Jeffries KA, Esmaeilniakooshkghazi A, Ogretmen B, Krupenko SA, Krupenko NI. CerS6 is a novel transcriptional target of p53 protein activated by non-genotoxic stress. J Biol Chem. 2016;291(32):1658696.

Article CAS PubMed PubMed Central Google Scholar

Yacoub A, Hamed HA, Allegood J, Mitchell C, Spiegel S, Lesniak MS, Ogretmen B, Dash R, Sarkar D, Broaddus WC, et al. PERK-dependent regulation of ceramide synthase 6 and thioredoxin play a key role in mda-7/IL-24-induced killing of primary human glioblastoma multiforme cells. Cancer Res. 2010;70(3):11209.

Article CAS PubMed PubMed Central Google Scholar

Eberle M, Ebel P, Wegner MS, Mannich J, Tafferner N, Ferreiros N, Birod K, Schreiber Y, Krishnamoorthy G, Willecke K, et al. Regulation of ceramide synthase 6 in a spontaneous experimental autoimmune encephalomyelitis model is sex dependent. Biochem Pharmacol. 2014;92(2):32635.

Article CAS PubMed Google Scholar

Bai X, He T, Liu M, Li L, Chen J, Cao M, Liu Y, Yang C, Jia W, Tao K, et al. Integrative analysis of MicroRNAs and mRNAs in LPS-induced macrophage inflammation based on adipose tissue stem cell therapy. Inflammation. 2021;44(1):40720.

Article CAS PubMed Google Scholar

Hernandez-Corbacho MJ, Canals D, Adada MM, Liu M, Senkal CE, Yi JK, Mao C, Luberto C, Hannun YA, Obeid LM. Tumor Necrosis Factor-alpha (TNFalpha)-induced ceramide generation via ceramide synthases regulates loss of Focal Adhesion Kinase (FAK) and programmed cell death. J Biol Chem. 2015;290(42):2535673.

Article CAS PubMed PubMed Central Google Scholar

Kim YR, Lee EJ, Shin KO, Kim MH, Pewzner-Jung Y, Lee YM, Park JW, Futerman AH, Park WJ. Hepatic triglyceride accumulation via endoplasmic reticulum stress-induced SREBP-1 activation is regulated by ceramide synthases. Exp Mol Med. 2019;51(11):116.

PubMed PubMed Central Google Scholar

Stiban J, Perera M. Very long chain ceramides interfere with C16-ceramide-induced channel formation: a plausible mechanism for regulating the initiation of intrinsic apoptosis. Biochim Biophys Acta. 2015;1848(2):5617.

Article CAS PubMed Google Scholar

Nicholson RJ, Pezzolesi MG, Summers SA. Rotten to the cortex: ceramide-mediated lipotoxicity in diabetic kidney disease. Front Endocrinol (Lausanne). 2020;11:622692.

Article PubMed Google Scholar

Mantovani A, Lunardi G, Bonapace S, Dugo C, Altomari A, Molon G, Conti A, Bovo C, Laaksonen R, Byrne CD, et al. Association between increased plasma ceramides and chronic kidney disease in patients with and without ischemic heart disease. Diabetes Metab. 2021;47(1):101152.

Article CAS PubMed Google Scholar

Mitsnefes M, Scherer PE, Friedman LA, Gordillo R, Furth S, Warady BA. Ceramides and cardiac function in children with chronic kidney disease. Pediatric Nephrol (Berlin, Germany). 2014;29(3):41522.

Article Google Scholar

Ichi I, Kamikawa C, Nakagawa T, Kobayashi K, Kataoka R, Nagata E, Kitamura Y, Nakazaki C, Matsura T, Kojo S. Neutral sphingomyelinase-induced ceramide accumulation by oxidative stress during carbon tetrachloride intoxication. Toxicology. 2009;261(12):3340.

Article CAS PubMed Google Scholar

Amsterdam EA, Wenger NK, Brindis RG, Casey DE Jr, Ganiats TG, Holmes DR Jr, Jaffe AS, Jneid H, Kelly RF, Kontos MC, et al. 2014 AHA/ACC guideline for the management of patients with Non-ST-elevation acute coronary syndromes: a report of the American College of Cardiology/American Heart Association task force on practice guidelines. J Am Coll Cardiol. 2014;64(24):e139228.

Article PubMed Google Scholar

OGara PT, Kushner FG, Ascheim DD, Casey DE Jr, Chung MK, de Lemos JA, Ettinger SM, Fang JC, Fesmire FM, Franklin BA, et al. 2013 ACCF/AHA guideline for the management of ST-elevation myocardial infarction: a report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol. 2013;61(4):e78140.

Article PubMed Google Scholar

Granger CB, Goldberg RJ, Dabbous O, Pieper KS, Eagle KA, Cannon CP, Van De Werf F, Avezum A, Goodman SG, Flather MD, et al. Predictors of hospital mortality in the global registry of acute coronary events. Arch Intern Med. 2003;163(19):234553.

Article PubMed Google Scholar

Gensini GG. A more meaningful scoring system for determining the severity of coronary heart disease. Am J Cardiol. 1983;51(3):606.

Article CAS PubMed Google Scholar

Jia Y, Gao Y, Li D, Cao Y, Cheng Y, Li F, Xiao L, Jiang Y, Wan Z, Zeng Z, et al. Geriatric nutritional risk index score predicts clinical outcome in patients with acute ST-segment elevation myocardial infarction. J Cardiovasc Nurs. 2020;35(6):E44-e52.

Article PubMed Google Scholar

Jia Y, Li H, Li D, Li F, Li Q, Jiang Y, Gao Y, Wan Z, Cao Y, Zeng Z, et al. Prognostic value of Braden scale in patients with acute myocardial infarction: from the retrospective multicenter study for early evaluation of acute chest pain. J Cardiovasc Nurs. 2020;35(6):E53-e61.

Article PubMed Google Scholar

Mehran R, Aymong ED, Nikolsky E, Lasic Z, Iakovou I, Fahy M, Mintz GS, Lansky AJ, Moses JW, Stone GW, et al. A simple risk score for prediction of contrast-induced nephropathy after percutaneous coronary intervention: development and initial validation. J Am Coll Cardiol. 2004;44(7):13939.

PubMed Google Scholar

Section 2: AKI Definition. Kidney international supplements 2012;2(1):1936. https://doi.org/10.1038/kisup.2011.32

Sarafian MH, Gaudin M, Lewis MR, Martin FP, Holmes E, Nicholson JK, Dumas ME. Objective set of criteria for optimization of sample preparation procedures for ultra-high throughput untargeted blood plasma lipid profiling by ultra performance liquid chromatography-mass spectrometry. Anal Chem. 2014;86(12):576674.

Article CAS PubMed Google Scholar

Huang Q, Hao S, Yao X, You J, Li X, Lai D, Han C, Schilling J, Hwa KY, Thyparambil S, et al. High-throughput quantitation of serological ceramides/dihydroceramides by LC/MS/MS: pregnancy baseline biomarkers and potential metabolic messengers. J Pharm Biomed Anal. 2021;192:113639.

Article CAS PubMed Google Scholar

Pickering JW, Blunt IRH, Than MP. Acute kidney injury and mortality prognosis in acute coronary syndrome patients: a meta-analysis. Nephrology (Carlton). 2018;23(3):23746.

Article PubMed Google Scholar

Yu J, Li D, Jia Y, Li F, Jiang Y, Zhang Q, Gao Y, Liao X, Zeng R, Wan Z. Nutritional risk screening 2002 was associated with acute kidney injury and mortality in patients with acute coronary syndrome: Insight from the REACP study. Nutr Metab Cardiovasc Dis. 2021;31(4):11218.

Article PubMed Google Scholar

Gencer B, Morrow DA, Braunwald E, Goodrich EL, Hilvo M, Kauhanen D, Sabatine MS, Laaksonen R, ODonoghue ML. Plasma ceramide and phospholipid-based risk score and the risk of cardiovascular death in patients after acute coronary syndrome. Eur J Prev Cardiol. 2020. https://doi.org/10.1093/eurjpc/zwaa143.

Article PubMed Google Scholar

Yao K, Wang Y, Xu D, Liu X, Shen C, Hu W, Wang Z, Wu R, Tang X, Sun A, et al. Effect of combined testing of ceramides with high-sensitive troponin T on the detection of acute coronary syndrome in patients with chest pain in China: a prospective observational study. BMJ Open. 2019;9(7):e028211.

Article PubMed PubMed Central Google Scholar

Abells-Sequeiros RA, Raposeiras-Roubn S, Abu-Assi E, Gonzlez-Salvado V, Iglesias-lvarez D, Redondo-Diguez A, Gonzlez-Ferreiro R, Ocaranza-Snchez R, Pea-Gil C, Garca-Acua JM, et al. Mehran contrast nephropathy risk score: Is it still useful 10 years later? J Cardiol. 2016;67(3):2627.

Article PubMed Google Scholar

Koowattanatianchai S, Chantadansuwan T, Kaladee A, Phinyo P, Patumanond J. Practical risk stratification score for prediction of contrast-induced nephropathy after primary percutaneous coronary intervention in patients with acute ST-segment elevation myocardial infarction. Cardiol Res. 2019;10(6):3507.

Article PubMed PubMed Central Google Scholar

Rodriguez F, Bonacasa B, Fenoy FJ, Salom MG. Reactive oxygen and nitrogen species in the renal ischemia/reperfusion injury. Curr Pharm Des. 2013;19(15):277694.

Article CAS PubMed Google Scholar

Ueda N, Camargo SMR, Hong X, Basnakian AG, Walker PD, Shah SV. Role of ceramide synthase in oxidant injury to renal tubular epithelial cells. J Am Soc Nephrol. 2001;12(11):238491.

Article CAS PubMed Google Scholar

Kalhorn T, Zager RA. Renal cortical ceramide patterns during ischemic and toxic injury: assessments by HPLC-mass spectrometry. Am J Physiol. 1999;277(5):F723-733.

CAS PubMed Google Scholar

Bergman BC, Brozinick JT, Strauss A, Bacon S, Kerege A, Bui HH, Sanders P, Siddall P, Kuo MS, Perreault L. Serum sphingolipids: relationships to insulin sensitivity and changes with exercise in humans. Am J Physiol Endocrinol Metab. 2015;309(4):E398-408.

Article CAS PubMed PubMed Central Google Scholar

Couto SMF, Machado DI, Conde C, Silva VC, Souza AA, Peres KB, Brandi BA, Vattimo MFF. Physical training is a potential modifier of risk for contrast-induced acute kidney injury in diabetes mellitus. Biomed Res Int. 2020;2020:1830934.

More here:
Long- and very long-chain ceramides are predictors of acute kidney ... - Cardiovascular Diabetology

New strategy positions B.C. as a global hub for life sciences | BC … – BC Gov News

Selina Robinson, Minister of Post-Secondary Education and Future Skills

Our life sciences sector is vital to creating a future of prosperity and innovation. Our Province is focused on preparing people for the jobs of tomorrow through our Future Ready plan, which will make education more accessible, affordable and relevant so we can build a stronger B.C. together.

Wendy Hurlburt, president and chief executive officer, Life Sciences BC

Building on the momentum of our life sciences thriving sector, this first-ever provincial life sciences strategy sets B.C. up to be a global leader in life sciences and will advance the health of British Columbians through the discovery and development of innovative products, solutions and services while diversifying and growing our economy by creating high-paying jobs.

Dr. Allen Eaves, president and chief executive officer, StemcellTechnologies

As Canadas largest biotechnology company, StemcellTechnologies is experiencing tremendous growth along with many other biotechs in B.C. All of us will require more homegrown research and biomanufacturing talent in the years ahead to remain internationally competitive.The B.C. governments Life Sciences and Biomanufacturing Strategy takes a thoughtful approach to strengthening the province from the perspective of both economic growth and health preparedness.

Bev Holmes, president and chief executive officer, Michael Smith Health Research BC

The new Life Sciences and Biomanufacturing Strategy further positions British Columbia as a leader in an area that is critical to peoplehere and around the world. The plan aligns with our work to support academic research and maximize benefits of clinical trials, which will create positive impacts on our economy, jobsand health.

Jennifer Figner, interim vice-president, academic and research, British Columbia Institute of Technology (BCIT)

BCITs mandate is to support the workforce development of the province, and we will play a major role in the training needsto ensure the future success and growth of the life sciences sector.With our partner, the Canadian Alliance for Skills and Training in Life Sciences, we look forward to welcominglearners to the National Biomanufacturing Training Centre.

Murray McCutcheon, senior vice-president, partnering, AbCellera

We believe that investments in life sciences talent and infrastructure are critical to building British Columbias ability to translate early scientific innovation into economic growth that makes our communities stronger today and tomorrow.

Suzanne Gill, president and chief executive officer, Genome BC

This new Life Sciences and Biomanufacturing Strategy will supercharge our already world-class life sciences sector and position us as a global leader in research and innovation, delivering new technologies and treatments that benefit B.C., Canadaand the world. Genome BC is proud to support this strategy by championing research and innovation to drive the responsible uptake of genomic technologies.

Cheryl Maitland, interim chief executive officer, Business Council of British Columbia

With an increasing number of scaling companies, highly skilled talent and research advancements, B.C.'s life sciences and biomanufacturing sector is asignificant source of employment, fuelling the growth of clean-tech businesses and contributing to our economy. The newly launched strategypresents a road map to build on these strengths and establish B.C. as a thriving global hubthat positively contributes to British Columbians health and economic well-being.

Deborah Buszard, interim president, University of British Columbia (UBC)

As B.C.s largest health-research and innovation organization and the lead institution for Canadas Immuno-Engineering and Biomanufacturing Hub, UBC welcomes the launch of the B.C. Life Sciences and Biomanufacturing Strategy and looks forward to continued collaboration with partners across academia, health care, industry, non-profits and government.Working together toward the strategys vision, we can create new opportunities for British Columbians and save more lives sooner.

Dr. Penny Ballem,member of the Council of Expert Advisors to the Government of Canada on biomanufacturing and life sciences

The life sciences strategy is an unprecedented opportunity for the remarkable life sciences sector in the province, our academic institutions and the health sector to leverage the investments being made by the federal and provincial government, and work together to benefit the lives of British Columbians and others across the country and globally, and support the biodiversity of our province and the planet.

More:
New strategy positions B.C. as a global hub for life sciences | BC ... - BC Gov News