header logo image


Page 9«..891011..2030..»

Archive for the ‘Immune System’ Category

New study could change what we eat to supercharge immune system and fight disease – WOODTV.com

Friday, August 19th, 2022

GRAND RAPIDS, Mich. (WOOD) The soldiers of our immune system were long thought to be fueled only by the foods we eat. However, researchers at Van Andel Institute believe the findings from their new study reveal T cells have a much wider appetite than originally thought.

Every process in the body is powered by metabolism, which in turn is fueled by the nutrients we consume through our diet, Russell Jones, Ph.D., chair of Van Andel Institutes Department of Metabolism and Nutritional Programming said. We found that immune cells are much more flexible in selecting the nutrient fuels they consume and, importantly, that they prefer some nutrients that were previously dismissed as waste. This understanding is crucial for optimizing T cell responses and developing new strategies for boosting our ability to fight off disease.

Jones, who is the co-author of the study published this week in Cell Metabolism, says the findings could create a path for personalized dietary recommendations that would supercharge immune cells and provide more effective therapies for cancer and other diseases.

Joneses research took a new approach to studyin T cells. In previous studies, the cells were grown in lab dishes with nutrient-contatining media. But Jones believed those nutrients were similar to a diet of eggs and toast. This time, Jones and his colleagues developed a more diverse sample of nutrients for the research and the outcome was much different.

We found that, when we offer them a full buffet, these cells actually prefer a wider array of fuels than previously believed, Jones said. This has major implications for how we tailor dietary recommendations as ways to promote health and combat disease.

Jones explains the research through what they discovered from lactate, a cellular waste that causes muscle aches and pains and a byproduct of cancer cells that allows the disease to attack other tissue and avoid the immune system. When the T cells were given the choice between glucose and lactate, they chose the lactate to power their energy production which enhanced their overall function.

According to VAI, there is research that suggests too much lactate is bad for T cells, but Jones work provides the idea that small amounts may increase their overall function.

Jones and his team plan to take their findings and use them to take a closer look at the unique connection between metabolism and the immune system to learn more about how they work together.

Hear from Dr. Jones below.

See the original post here:
New study could change what we eat to supercharge immune system and fight disease - WOODTV.com

Read More...

Your Immune System Will Thrive With This Elderberry Hill Liquid Morning Multivitamin – Men’s Journal

Friday, August 19th, 2022

Mens Journal aims to feature only the best products and services. We update when possible, but deals expire and prices can change. If you buy something via one of our links, we may earn a commission.Questions? Reach us at shop@mensjournal.com.

Summers almost over guys. That means the Fall is right around the corner and the temps are gonna drop. Were about to enter the cold season. No one wants to deal with the cold, or anything even worse than that. We need to boost our immune systems in any way we can. And the Elderberry Hill Liquid Morning Multivitamin will be a big help.

The Elderberry Hill Liquid Morning Multivitamin is going to be a big help because of all the ingredients that each spoonful is chock full of. You got a lot of goodies in here that are perfect for getting that immune system going stronger than before. Ingredients thatll absorb into the bloodstream better because of its liquid form.

Whats in the Elderberry Hill Liquid Morning Multivitamin? Vitamins A, C, D3, E, Thiamin, Zinc, and all sorts of other goodies. All of which form together to not just help your immune system, but also help with the health of your hair, skin, and nails, as well as boost your energy levels. How can you beat that?

Even better is that this vitamin tastes pretty damn good too. Its almost like a little treat for yourself to get your morning started just right. And there are no sugars or GMOs in here. Its gluten and nut-free, so everyone, including vegans, can really enjoy this vitamin on the quest to boost your immune system.

Before the Fall comes along and brings the chill with it, we think you guys absolutely need to pick up the Elderberry Hill Liquid Morning Multivitamin. Its going to go down nice and smooth, getting your body prepped for the incoming season. Pick up a bottle now and get the prep started early. You wont regret it.

Get It: Pick up the Elderberry Hill Liquid Morning Multivitamin ($35) at Amazon

Check out the Daily Deals over at Amazon

For any questions or concerns you have about the Coronavirus, head on over to the CDC

Check out the great products and gear we recommend to Mens Journal readers

Related Links

The 4 Best Multivitamin For Men: Support Health and Performance

Look And Feel 10 Pounds Lighter With A Best Selling Colon Detox

6 Exogenous Ketone Supplements to Keep You Losing Weight In Ketosis

Visit link:
Your Immune System Will Thrive With This Elderberry Hill Liquid Morning Multivitamin - Men's Journal

Read More...

Extending dogs’ lives, and sex and the immune system – MIT Technology Review

Friday, August 19th, 2022

Matt Kaeberlein is what you might call a dog person. He has grown up with dogs and describes his German shepherd, Dobby, as really special. But Dobby is 14 years oldaround 98 in dog years. Im very much seeing the aging process in him, says Kaeberlein, who studies aging at the University of Washington in Seattle.

Kaeberlein is co-director of the Dog Aging Project, an ambitious research effort to track the aging process of tens of thousands of companion dogs across the US. He is one of a handful of scientists on a mission to improve, delay, and possibly reverse that process to help them live longer, healthier lives.

But dogs are just the beginning. Because theyre a great model for humans, anti-aging or lifespan-extending drugs that work for dogs could eventually benefit people, too. In the meantime, attempts to prolong the life of pet dogs can help people get onboard with the idea of life extension in humans. Read the full story.

Jessica Hamzelou

The quest to show that biological sex matters in the immune system

For years, microbiologist Sabra Klein has painstakingly made the case that sexdefined by biological attributes such as our sex chromosomes, sex hormones, and reproductive tissuescan influence immune responses.

Through research in animal models and humans, Klein and others have shown how and why male and female immune systems respond differently to the flu virus, HIV, and certain cancer therapies, and why most women receive greater protection from vaccines but are also more likely to get severe asthma and autoimmune disorders (something that had been known but not attributed specifically to immune differences.)

In the 1990s, scientists often attributed such differences to gender rather than sexto norms, roles, relationships, behaviors, and other sociocultural factors as opposed to biological differences in the immune system. Klein has helped spearhead a shift in immunology, a field that long thought sex differences didnt matterand shes set her sights on pushing the field of sex differences even futher. Read the full story.

View original post here:
Extending dogs' lives, and sex and the immune system - MIT Technology Review

Read More...

Sure Signs Your Immune System Isn’t as Strong as it Should Be Eat This Not That – Eat This, Not That

Friday, August 19th, 2022

Prioritizing your health has never been more important. As COVID cases continue to spike and the monkeypox outbreak has now become a public health emergency, having a strong immune system is essential. Daily habits can impact our immune health and lifestyle choices such as smoking, poor diet, and too much alcohol consumption can weaken your immunity. But there are ways to help strengthen our body and knowing the signs of a troubled immune system is a start. Eat This, Not That! Health spoke with Dr. Tom Yadegar, Pulmonologist and Medical Director of the Intensive Care Unit at Providence Cedars-Sinai Tarzana Medical Center who shares what to know about your immune system and warning signals it's not as healthy as it should be. Read onand to ensure your health and the health of others, don't miss these Sure Signs You've Already Had COVID.

Dr. Yadegar states, "The immune system is the defender of the body. Composed of two arms, innate and adaptive, the innate system is a nonspecific response that fights any type of foreign invader that comes into contact with the body. This arm is generally the same in most people and is composed of white cells. The second arm, known as the adaptive immune system, is specific to the foreign invader and targets it using antibodies from previous infections or vaccines."

"When exposed to a foreign invader, the immune system creates antibodies in response to prevent severe symptoms in case of repeated exposure," says Dr. Yadegar. "When this process turns against healthy tissue instead of foreign pathogens, the immune system attacks the body, leading to an autoimmune state."

Dr. Yadegar shares, "Getting proper sleep, nutrition and regular exercise is a hallmark to keeping the immune system functional. Adequate vitamin intake, including vitamin C and vitamin D, is also important in ensuring a strong immune system. Patients who may have immunodeficiency, such as IgG deficiency, can also receive infusions to help keep their immune system healthy."

Dr. Yadegar tells us, "Fighting infections requires a lot of energy. When the body is depleted of its normal energy level, the immune system is weakened and can become susceptible to opportunistic infections. People generally feel this when they are tired. Ensuring a schedule of restful sleep, eating a balanced diet with fruits and vegetables and drinking enough water helps ensure your immune system is ready to answer the call of an infection."6254a4d1642c605c54bf1cab17d50f1e

According to Dr. Yadegar, "Infections that require multiple courses of antibiotics within a year may be a sign of a weakened immune system that is not able to fight off pathogens. Patients should be evaluated by their healthcare provider in order to further investigate the underlying cause."

"Normal wounds require the immune system to bring nutrients to repair damaged tissue," says Dr. Yadegar. "When this process is compromised, wounds are unable to heal properly, which signals a slow immune system. Delayed wound healing is indicative of a poorly-functional immune system, and should be evaluated by a healthcare provider."

Dr. Yadegar explains, "Long-term stress compromises the body's natural immunity, which can lead to higher risk of infections. While stress is inevitable in our fast-paced lives, taking steps to mediate stress can help. Whether meditation, exercise, or deep-breathing, it's important to tailor stress-relief to the individual in order to best improve their stress levels."

Heather Newgen

The rest is here:
Sure Signs Your Immune System Isn't as Strong as it Should Be Eat This Not That - Eat This, Not That

Read More...

Can the monkeypox vaccine stop the current outbreak? : Goats and Soda – NPR

Friday, August 19th, 2022

A health-care worker prepares to administer a free monkeypox vaccine in Wilton Manors, Florida. The question: Can vaccination slow the outbreak? Joe Raedle/Getty Images hide caption

A health-care worker prepares to administer a free monkeypox vaccine in Wilton Manors, Florida. The question: Can vaccination slow the outbreak?

Finally, we have a glimmer of good news about monkeypox: The outbreaks in some countries, including the U.K., Germany and parts of Canada, are starting to slow down.

On top of that, the outbreak in New York City may also be peaking and on the decline, according to new data from the city's health department.

All these outbreaks are "far from extinguished," says infectious disease specialist Dr. Donald Vinh at McGill University in Montreal. But there are signs that, in some places, "they're a bit more under control than they had been."

For example, in the U.K., the number of new cases reported each day has steadily declined since late July, dropping from 50 daily cases to only about 25. (By contrast, here in the U.S., daily cases are still increasing. Since late July, the U.S. daily count has risen from 350 new cases to 450 cases.)

Some health officials credit the monkeypox vaccine and its quick rollout as the key factor that's slowing the spread of the virus in the U.K..

"Over 25,000 have been vaccinated with the smallpox vaccine, as part of the strategy to contain the monkeypox outbreak in the UK.," the U.K. Health Security Agency wrote on Twitter on Tuesday. "These 1000s of vaccines, given by the NHS to those at highest risk of exposure, should have a significant impact on the transmission of the virus."

Indeed, the U.K. and parts of Canada rolled out the vaccine in late May, weeks before doses became available in most U.S. cities.

But does the monkeypox vaccine have the ability to stop or curb the spread of the virus? To answer that question, we need to first understand a few basics about this vaccine.

What actually is the monkeypox vaccine? How does it work?

So the monkeypox vaccine is actually the smallpox vaccine. Maybe that sounds a bit strange, but in fact the two pox viruses are related. They're a bit like cousins.

Health-care workers used an earlier version of this vaccine to eradicate smallpox in the 1970s. So versions of this vaccine have been given to hundreds of millions of people over the past century. It has a long track record.

Back in the late 1980s, researchers started to notice something remarkable about this vaccine. During a monkeypox outbreak in the Democratic Republic of the Congo (then called Zaire), people who were immunized against smallpox were less likely to get monkeypox. They were protected. And not by just a little but by quite a bit. In a small study, published in 1988, researchers estimated the smallpox vaccine offered about 85% protection against monkeypox.

Now, the virus in this study was a different variant of monkeypox than the one circulating in the current international outbreak and that variant wasn't spreading primarily through sexual contact, as monkeypox is doing today. So we don't know how well these findings will translate to protection during the current outbreak. Which brings us to the next question.

How well does the vaccine protect against a monkeypox infection?

The short answer is: "We don't know," says infectious disease specialist Dr. Boghuma Titanji at Emory University.

There's no doubt the vaccine will offer some protection, Titanji says. "But right now, we still need studies in people to understand what level that protection actually is."

In North America and Europe, countries are primarily rolling out a vaccine called JYNNEOS, which was developed in the early 21st century. The goal with this vaccine is to increase its safety compared to the older vaccine, whose life-threatening complications, including encephalitis and skin necrosis, occurred in about 4 out of every million people vaccinated. That vaccine also could cause damaging skin lesions in people with eczema or weakened immune systems. (Note: There is a shortage of the JYNNEOS vaccine, and no doses have been shared with or sold to countries in Africa, which have experienced monkeypox outbreaks since the 1970s.)

Although older versions of the vaccine have been tested thoroughly in people, there has never been a large, clinical study to measure JYNNEOS's ability to protect against a monkeypox infection in people or to stop transmission of the virus.

What is known about the vaccine, in terms of its efficacy against monkeypox, comes from studies in macaques, and immunological studies in people, which demonstrated the vaccine triggers the production of monkeypox antibodies in people's blood.

"So we know that the vaccine does stimulate the immune system and people produce antibodies when they receive the vaccine," Titanji says, "but we don't have a clinical data in humans to actually tell us, 'Okay, that immune response translates to this level of protection against getting infected with monkeypox or reducing the severity of monkeypox disease if you do get infected.' "

And it's not a guarantee of protection. In this current outbreak, scientists have already begun to document breakthrough infection with this vaccine, the World Health Organization reported Thursday. "[This] is also really important information because it tells us that the vaccine is not 100% effective in any given circumstance," said Dr. Rosamund Lewis of WHO. "We cannot expect 100% effectiveness at the moment based on this emerging information."

And so when Titanji gives a person the JYNNEOS vaccine at her clinic, she is very clear about what the vaccine can and can't do. "I tell them, 'We do know that you're going to get some protection from this vaccine. Some protection is better than no protection. We also do know that the vaccine can reduce the severity of the disease if you do get infected. But we don't know for a fact that you would be completely protected from getting monkeypox.' "

Can this vaccine if given to the people who need it the most slow down the outbreak?

So the new data from the U.K. and Germany suggest that indeed this vaccine can curb the spread of monkeypox.

But Dr. Vinh at McGill University says it's way too soon to say the vaccine, alone, is the only factor contributing to the slow down in these countries. "No single measure is going to really be the solution here," says Vinh.

In addition to vaccination, people at high risk need to learn how they can protect themselves. And doctors have to learn how to spot monkeypox cases, he says.

Right now the percentage of monkeypox tests coming back positive is still incredibly high, Titanji says. "The positivity rate is close to 40%." And that means doctors are missing many cases. Specifically, they are still mistaking monkeypox for other sexually transmitted diseases such as syphyllis.

"I can tell you, from the lens of a clinician, that monkeypox is very, very easy to mistake for another infectious disease," she says.

Some people have had to visit clinics two or three times and even have been treated for another STD before the clinician suspects monkeypox.

"You really have to maintain a very high index of suspicion because some of the lesions are so subtle and the clinical presentation is so variable," she says. "At this phase of the outbreak, we should be over testing rather than under testing. If a doctor even remotely suspects monkeypox, they should be sending a test for it."

Otherwise people can't receive treatment for monkeypox and they can unknowingly spread it to others. And the outbreak will continue to grow while people wait to receive a vaccine and for that vaccine to begin working.

Visit link:
Can the monkeypox vaccine stop the current outbreak? : Goats and Soda - NPR

Read More...

Getting A Grip On Influenza: The Pursuit Of A Universal Vaccine (Part 2) – Forbes

Friday, August 19th, 2022

Taiwan researchers sort through eggs used for the cultivation of swine flu vaccine, in a plant in ... [+] Taichung, on June 18, 2009. Taiwan is set to mass produce swine flu vaccine in October, as the island's confirmed cases rose to 58 as of June 17. AFP PHOTO/PATRICK LIN (Photo credit should read PATRICK LIN/AFP via Getty Images)

This is a short series about a recent breakthrough on the road to developing a much sought-after broadly neutralizing vaccine against all influenza A viruses. If successful, it may act as a precursor to a truly universal flu vaccine, one that protects against all types, subtypes, and lineages of the virus. The breakthrough may also provide a blueprint for developing a Covid-19 vaccine that retains its efficacy in the face of new variants.

In the first part of this series, I gave a brief overview of the history and nature of influenza viruses, including why it has been so difficult to develop successful vaccines. The next few articles discuss some of the attempts that have been made to overcome these challenges, including their shortcomings. And in the last installments, I will offer a detailed analysis of the latest and most promising advances in the field.

The Seasonal Approach

Picking up where we left off in the previous article, any successful influenza vaccine has to account for the ability of influenza viruses to mutate. Genetic mutations to vital proteins can lead to antigenic variation changes to parts of the virus that our immune system relies on to stimulate its memory. Although various different parts of the virus serve as antigens, the surface proteins that help it enter and exit host cells are some of the most important. Changes to these proteins can prevent our antibodies from recognizing the virus, rendering them unable to block its spread. Antigenic variation is responsible for influenza reinfections, leading to seasonal flu outbreaks.

In an attempt to circumvent the issue of antigenic variation, vaccine manufacturers update the flu shot each year based on the latest circulating influenza strains. The idea is to expose our immune system to the antigens it is most likely to encounter during flu season, helping it to build up its antigen-specific defenses in advance once our immune system has built up its memory, it can jump into action straight away should we become infected.

Which influenza strains ultimately get used to make the yearly flu shot is decided on the basis of data collected throughout the year by the World Health Organizations (WHO) Global Influenza Surveillance and Response System (GISRS). This surveillance and response system is made up of roughly 150 different laboratories spread across the globe, each of which gathers thousands of influenza samples from sick patients. The most prevalent viral strains are then shared with five WHO Collaborating Centers for Influenza, which perform further analysis. Two times a year once in preparation for flu season in the Northern Hemisphere, and another in preparation for flu season in the Southern Hemisphere Directors of the WHO Collaborating Centers, Essential Regulatory Laboratories, and representatives of a few of the smaller national laboratories come together to: review the results of surveillance, laboratory, and clinical studies, and the availability of flu vaccine viruses and make recommendations on the composition of flu vaccines. Once the WHO vaccine composition committee has made its recommendations, each country makes a final decision on which viruses they will choose to use in their flu vaccines.

In the United States, all influenza vaccines are quadrivalent, meaning they contain four different influenza viruses. This is done to broaden protection against the various influenza subtypes and lineages known to drive seasonal outbreaks: influenza A (H1N1), influenza A (H3N2), influenza B/Victoria, and influenza B/Yamagata. Quadrivalent vaccines will also protect against any other influenza viruses that are antigenically similar.

Although this may seem like a relatively reliable process, there is one glaring drawback to the seasonal vaccination approach: vaccines produced in this way are nowhere near as effective as we might hope. At best, they protect 60% of people from illness, but this number can, and often does, drop much lower. For the influenza A (H3N2) subtype, vaccine effectiveness hovers around 33%. Of course, any protection is better than no protection, but it is still suboptimal remember, these numbers represent best case scenarios, years where the viruses selected for use in vaccines are well matched to those that actually end up circulating during the flu season. So, where are things going wrong?

Missing the Target: Egg-based Vaccines

Selection of candidate vaccine viruses (CVVs) is only one part of the equation, growing them is another. This is no simple feat considering they need to be available in bulk, enough to make millions of vaccines. For the past 70 years, the majority of manufacturers have turned to chicken eggs in order to achieve the necessary growth (Figure 1). The candidate vaccine viruses are injected into fertilized hens eggs and left to incubate for a few days. During this period, the viruses are able to replicate. The fluid in the eggs is then extracted and the viruses are killed (inactivated). Finally, the antigen of choice usually the hemagglutinin surface protein is isolated from the killed viruses and purified, making it ready for use in vaccines. Even now, most flu vaccines continue to be egg-based.

FIGURE 1. An overview of the steps involved in producing egg-based vaccines.

But there are two issues with this approach. First, growing the viruses in eggs is a fairly slow process. This means the selection of candidate vaccine viruses has to happen far in advance of flu season, to make sure manufacturers have enough time to produce the amounts needed. In the six to nine months it takes to grow and purify enough virus, the wild type influenza strains continue to mutate and change. If these changes impact the antigen, the wild type viruses may escape the immunity that the vaccines provide us, reducing their effectiveness. When this happens, the viruses are referred to as escape mutants.

A growing body of research suggests that a second factor may be even more important: egg-adapted changes. Because the candidate vaccine viruses are human influenza viruses, growing them in chicken eggs carries the risk that they adapt to the new immune niche while replicating. The immune niche of chickens is different to that of humans, so adaptations that improve viral fitness in chickens may result in genetic and antigenic changes to the viruses. As before, these changes can lead to a drop in vaccine effectiveness, since the vaccine strains no longer resemble the circulating wild type strains; the egg-adapted vaccines end up training our immune system to recognize the wrong viruses, thus hampering its ability to respond efficiently come flu season.

Egg Substitutes: New Ways of Growing Candidate Viruses

In response to these issues, manufacturers have tried to develop new production methods that avoid using chicken eggs to culture candidate viruses. This search has led to a cell-based approach and a recombinant approach (Figure 2).

FIGURE 2. Timeline of current influenza vaccine production methods. Schematic overview of egg-based, ... [+] cell-based and protein-based (recombinant) influenza vaccine production.

Cell-based vaccines are produced using candidate viruses grown in mammalian cells rather than chicken eggs. Aside from this, the manufacturing process between the two is virtually identical: candidate vaccine viruses are grown in mammalian cell cultures by the CDC, these are then handed over to private manufacturers who inoculate the viruses into mammalian cells, the viruses are left to replicate for a few days before being harvested, and finally, purified. Although approved in 2012, it wasnt until this past 2021-2022 flu season that fully egg-free, cell-based vaccines were produced previously, the initial production of candidate vaccine viruses by the CDC was still done using fertilized hens eggs, and only after being handed over to the private sector were the viruses mass-produced in mammalian cells.

Using the cell-based approach eliminates egg-adapted changes in candidate viruses, keeping the viruses as close as possible to the wild type influenza strains predicted to circulate during flu season. An added benefit of cell-based vaccines is that the production process can be scaled up more quickly; mammalian cells can be frozen in advance to ensure steady supply, which could prove especially useful during pandemic outbreaks.

In theory, the lack of egg-adapted changes should improve vaccine effectiveness. But what about in practice? Although there still hasnt been enough research for a clear consensus to develop, initial findings suggest the difference in effectiveness is modest at best, and statistically insignificant at worst. This hints that egg-adapted changes might not play as important of a role as initially suspected; low vaccine efficacy can occur even when eggs are not used in the manufacturing process. That said, the 2021-2022 flu season marks the first time truly egg-free cell-based vaccines in which all four viruses are derived entirely through cell-based methods were used, so perhaps future research will yield different results. For now, things dont look too promising.

Recombinant vaccines provide a third option, and manage to overcome a crucial issue faced by the other two options: the lengthy, tedious virus production process. Whereas egg- and cell-based vaccines depend on candidate virus samples, recombinant manufacturing skips this step. Instead, recombinant vaccines are made by isolating the gene that makes the hemagglutinin surface protein from a wild type influenza virus. Once isolated, this gene is combined with a different kind of virus, called baculovirus. The new virus is known as a recombinant baculovirus and it is used to ferry the gene that makes the hemagglutinin antigen into a host cell line. As soon as the gene enters the cells, they begin to mass produce the hemagglutinin antigen. The antigen can then be extracted and purified before being assembled into a vaccine.

Given that they are entirely egg-free and dont require candidate virus samples, recombinant vaccines bypass the issue of egg-dependent changes. Due to the speed of production, there is also a decreased risk of escape mutants developing. As before, there is a paucity of comparative research, making it difficult to draw any firm conclusions, but early findings suggest recombinant vaccines may be more effective than traditional egg-based and cell-based vaccines, including improved antibody production.

Takeaway

Developing consistently protective influenza vaccines has proven difficult, with effectiveness frequently hovering somewhere between 40 and 60%. Too low, considering the threat posed by influenza.

A big part of the challenge is the mutability of the virus; it is constantly changing, making it hard for our immune system to keep up and retain useful memories of previous encounters. In response, public health agencies and scientists around the world develop new vaccines every year that prime our immune systems for the latest circulating strains. Sometimes scientists miss the mark with their predictions, in which case the circulating influenza strains do not match up with those in the vaccine, undermining vaccine effectiveness. At other times predictions are right on the money, but the vaccine production process impairs effectiveness either by being too slow and giving the wild type viruses time to mutate again, or because of mutations to the candidate vaccine strains during mass-production in chicken eggs.

Cell-based and recombinant vaccines aim to resolve the issues on the production side of things. The former by skipping the need for eggs, and by extension, the threat of egg-adapted changes. The latter by skipping the need for eggs as well as cutting down the time it takes to produce the vaccines, reducing the risk of escape mutants. Despite these advances, vaccine effectiveness has not yet seen the boost it needs.

The below table gives a summary of the advantages and disadvantages associated with these three production processes.

FIGURE 3. Advantages and disadvantages of strategies for influenza virus vaccine production.

The next article in this series will look at two additional technologies: intranasal vaccines and mRNA vaccines. Might they succeed where the more traditional strategies have wavered?

Continue reading here:
Getting A Grip On Influenza: The Pursuit Of A Universal Vaccine (Part 2) - Forbes

Read More...

Bells Are Ringing! How Immunotherapy is Unlocking Doors to a Cancer Cure – Georgetowner

Friday, August 19th, 2022

Your immune system is on a mission, constantly assessing threats, identifying invaders, and neutralizing or killing them off. It is a finely tuned network of organs, cells, proteins, and chemicals engaged in an existential battle. It asks the question: is this me or is this not me? And if its not me, what is it? Friend or foe?

Without the immune system, which has been honed and refined throughout the millennia of our existence as a species, we could not survive.

Samir N. Khleif, M.D., is also on a mission: to outsmart and disable cancer by overcoming its ability to evade or tolerate immunotherapeutic approaches.

Dr. Khleif, a practicing medical oncologist, a Biomedical Scholar, and professor, is the director of the Center for Immunology and Immunotherapy and the Loop Immuno-Oncology Research Laboratory at the Lombardi Comprehensive Cancer Center at Medstar Georgetown University Hospital. He and his team of assistant professors, post docs, research assistants, graduate students, and trainees focus on understanding how the immune system works, delineating the mechanisms of immune response and resistance to immunotherapy and re-engineering the immune cells with the goal of developing novel immune therapeutics.

Khleif is a long-time pioneer in cancer immunotherapy. Before joining Georgetown, he served as Director of Georgia Cancer Center, Augusta University, where he oversaw the development of a large integrated program focused on immunology, inflammation, tolerance basic science, and immune therapy. He also led the Cancer Vaccine Section, a nationally active Immune Therapy Program at the National Institutes of Health-National Cancer Institute, where he was one of the early pioneers of cancer vaccines and led some of the clinical trials. (Fun fact: Moderna and BioNTech, the names behind the mRNA technology now used to protect us against COVID, started out as cancer vaccine companies.)

He currently holds numerous patents and has published several important studies unraveling the understanding of the interaction of immune cells and cancer and on the mechanisms of tumor-induced suppression and the strategies used to overcome them. His research team has also developed models to understand how different kinds of immune therapies can be combined to work synergistically and he translated these findings into clinical trials with the intention of more widespread use.

We recently met with him in his lab to learn more about immunology a subject weve all come to know since the pandemic and to discuss his research. For all his stellar achievements and fierce intellect, he was a gracious host and a passionate teacher. He is also, we later learned, a painter and a musician who plays keyboard, saxophone, piano, and the violin, amateurly, he insists. His top scientist of all time is Albert Einstein and his favorite D.C. restaurant is Komi.

Commenting on the upcoming BellRinger Ride benefit for Lombardi, Khleif sees similarities between his life mission and bicycling: both activities have an anticipation to reach the end goal along with hard work and a sense of exploration or adventure. To find out what BellRingers all about, see our sidebar in our print edition here.

Born in Syria to Palestinian refugee parents, Khleif attended college and medical school in Jordan after spending seven weeks in Vermont to learn English. Although he originally wanted to be a physicist, his father swayed him into medicine where a love of research led him to the study of virology, molecular biology, vaccines and, now, his work in harnessing the power of immune system to disable cancer cell growth and proliferation.

For Khleif, the joy of discovery is the catalyst for his work. The more discoveries you find, he says, the more addicted you get. I tell my team: when you discover something, ask yourself, why did nature create like this? Why does it exist? Can we recreate it when its missing? How can we use this as a tool for therapy?

Khleif and his team concentrate on four main areas of research: tumor immunology and immunotherapeutics (unraveling the mechanism through which the immune system and cancer cells interact ); T-cell plasticity (how T-cells, a type of immune cell, can be re-reengineered to amp up their immune response); immunotherapeutic resistance (how and why tumors learn to override the patients natural defenses and therefore become unresponsive to immuno- and other therapies); and combination immunotherapy design (identifying the best combination of immunotherapeutics to enhance the best clinical response).

Interestingly, the lab is also studying how some natural products, such as vitamin C and selenium, can be used to boost immunity, reprogram and repair immune cells, and reverse the damage that cancer causes on the immune system. So, stock up on your fruits, vegetables and seafood.

In his other life as an advocate for global health and impact-driven healthcare, he led the development and served as the founding CEO of the King Hussein Cancer Center in Amman, Jordan, the regional cancer center in the Middle East. He also led the planning and development of cancer care projects in low-income countries dedicated to bringing cancer education, research, and treatment to underserved areas around the world.

Every day, as your immune system conducts its intricate surveillance, it is working to dispatch dangers before they become serious health risks. Dangers like an errant cell that may grow into cancer. With immunotherapy in their arsenal, Khleif and his team are unlocking new strategies to dethrone the emperor of all maladies and save, he estimates, millions of lives.

To learn more about Dr. Khleif, his research, patents, and publications go to: https://gufaculty360.georgetown.edu/s/contact/00336000019h06bAAA/samir-khleif. You can also view his patient-oriented video on immunotherapy here: https://youtu.be/afdq8Op-jQM

For a highly accessible and entertaining resource on the immune system, check out Philipp Dettmers Immune, https://youtu.be/afdq8Op-jQM. If you or a family member have been diagnosed with cancer and would like to better understand immunotherapy, visit the Cancer Support Community here: https://www.cancersupportcommunity.org/immunotherapy-cancer-it-right-you .

And to support innovative cancer research at the Georgetown Lombardi Comprehensive Cancer Center, join the inaugural BellRinger Bike Ride on Oct. 22. Donate or learn more at bellringer.org.

Continue reading here:
Bells Are Ringing! How Immunotherapy is Unlocking Doors to a Cancer Cure - Georgetowner

Read More...

Have The Immune System Booster Through the Dietary Supplements Only from Berkeley Immune Support Formula – Digital Journal

Friday, August 19th, 2022

Berkeley Immune Support Formula provides supplements to the diet that can help in the boosting of the immune system and thus be and remain physically stronger.

Los Angeles, CA (August 13, 2022) The immune system is an essential part of the human body as it works as the shield that protects it from different kinds of diseases. Berkeley Immune Support Formula highly recommends one to be immunoreactive and suggests some of the daily care that one can take as a measure to keep a check on it, such as checking the vitamin D, keeping control of weight, and having an immune support booster, and others.

The product of the organization is mainly derived from many vegetables such as broccoli and cabbage due to which the organization also recommends that one can intake those in other forms as well. As their product is composed of more such elements rich in the DIM, it promises to give a better immune system result and prove itself the best immune support booster.

The product has been designed to aid the cause of raising funds for nature-based biomedical research. The product is said to be rich in Selenium, Zinc, Sulforaphane, Lycopene, Zeaxanthin, Lutein, Citrus Bioflavonoids, and Vitamin D3. The manufacturers of the product have many experiences with the DIM substance. The sulforaphane that is delivered by the immune support booster product is said to be specially manufactured by the scientists for the product.

Through the Berkeley Formula product, the organization makes a promise to the customers to provide the nutrients, which will be equal to a bowl of salad. The product is said to be the first of its kind that is composed of a mixture of bioflavonoids and phytonutrients. The product supplied by the organization is said to be manufactured for the most bioavailability, and it is also said to be giving bioactive quantities of the DIM to the consumers.

The Berkeley Formula product is regarded as a very effective bioavailable supplement of the DIM and immune system booster by many doctors and scientists. In fact, they have been found to do more research on the products along with the addition of other nutrients. The product is also said to be very effective for sportspeople, relieving stress from work or school, pollution of the environment, the process of aging, and sleep deprivation as an immune system booster.

About Berkeley Immune Support Formula

The organization was founded by Dr. Leonard Bjeldanes, Dr. Gary Firestone, Dr. Christopher Benz, Dr. Giuseppe Del Priore, and Dr. Bob Eghbalieh. These doctors have a specialty in the product supplied by the organization which is the DIM supplements, and in clinical research. The main purpose of this research is to contribute to the medical field with the help of their products. The main goal of the organization is to help people lead a healthy life, via the development of top-class nutrition goods. The two divisions of the organization function, that are the nutritional sciences and the biopharmaceuticals functions in the marketing of their product and in the development of the different types of cures for deadly diseases by boosting the immune system of the body.

For more information about the services of the company and knowing the offers, please visit https://www.berkeleyformula.com/

Media Contact:

Berkeley Immune Support Formula

1434 Westwood Blvd. Suite #5, LA, CA 90024

Phone: 877-777-0719

Email: [emailprotected]

The rest is here:
Have The Immune System Booster Through the Dietary Supplements Only from Berkeley Immune Support Formula - Digital Journal

Read More...

COVID-19 and Flu Viruses Often Have a Deadly Accomplice: Bacterial Infections – Tufts Now

Friday, August 19th, 2022

The1918 influenza pandemicresulted in the loss of over 3% of the worlds population at least 50 million people. But it wasnt the flu virus that caused the majority of these deaths.

Ananalysis of lung samplescollected during that flu pandemic indicated that most of the deaths were likely due to bacterial pneumonia, which ran rampant in the absence of antibiotics. Even in more recent history, like the1957 H2N2and2009 H1N1flu pandemics, nearly 18% of patients with viral pneumonia had additional bacterial infections that increased their risk of death. And theCOVID-19 pandemicis no different.

With yet another flu season fast approaching in the midst of the ongoing COVID-19 pandemic, lessening the harm caused by these viruses is important to prevent deaths and reduce infections. However, many deaths associated with the flu and COVID-19 dont occur at the hand of the virus alone. Instead, its asecondary bacterial infectionthat is often at the root of the devastating consequences attributed to an initial viral infection.

I am animmunologistwho studies why and how cells die during bacterial and viral infections. Understanding the synergy between these microbes is critical not only for effective diagnosis and treatment, but also for managing current pandemics and preventing future ones. My colleagues and Ipublished a studyshowing how an immune system protein crucial to fighting against viruses also plays an indispensable role in fighting bacteria.

Multiple pathogens can cause multiple infections in different ways. Scientists distinguish each typebased on the timingof when each infection occurs.Coinfectionrefers to two or more different pathogens causing infections at the same time.Secondary or superinfections, on the other hand, refer to sequential infections that occur after an initial infection. Theyre often caused by pathogens resistant to antibiotics used to treat the primary infection.

How viral and bacterial infections interact with each other increases the potential harm they can cause. Viral respiratory infections can increase the likelihood of bacterial infections and lead to worse disease. The reason why this happens is often multifaceted.

Within your respiratory tract, the epithelial cells lining your airways and lungs serve as the first line of defense against inhaled pathogens and debris. However,viruses can kill these cellsand disrupt this protective barrier, allowing inhaled bacteria to invade. They can alsochange the surface of epithelial cellsto make them easier for bacteria to attach to.

Viruses can also alter the surface ofepithelial and immune cellsbyreducing the number of receptorsthat help these cells recognize and mount a response against pathogens. This reduction means fewer immune cells report to the viral infection site, giving bacteria an opening to launch another infection.

Patients who have a bacterial infection at the same time theyre battling the seasonal flu are more likely to wind up in a hospital.Nearly a quarterof patients admitted to the ICU with severe influenza also have a bacterial infection. One study on the 2010 to 2018 flu seasons found thatnearly 20% of patientsadmitted to the hospital with flu-associated pneumonia had acquired bacterial infections.

Another studyof patients hospitalized with viral or bacterial infections found that nearly half had a coinfection with another pathogen. These patients also had nearly double the risk of dying within 30 days compared to those with only a single infection.

Interestingly, thetwo bacteria speciesmost commonly involved in coinfections with the influenza virus areStreptococcus pneumoniaeandStaphylococcus aureus, which normally exist in the respiratory tract without causing disease. However, the influenza virus can damage the cell barrier of the lungs and disrupt immune function enough to make patients susceptible to infection by these otherwise benign bacteria.

Secondary bacterial infections are also exacerbating the COVID-19 pandemic. A 2021 review estimated that16% to 28% of adultshospitalized for COVID-19 also had a bacterial infection. These patients stayed in the hospital for twice as long, were four times more likely to need mechanical ventilation and had three times greater odds of dying compared to patients with only COVID-19.

The immune systemresponds differentlyto viruses and bacteria.Antiviralsdont work on bacteria, and antibiotics dont work on viruses. A better understanding of what pathways the body uses to regulate both antiviral and antibacterial infections is critical to addressing secondary and coinfections.

Recent workby my colleagues and me may provide a clue. Wesequenced the RNAof one type of immune cell, macrophages, in mice to identify what molecules were present in cells that were either protected from or died due to bacterial infection.

We identifiedZ-DNA binding protein (ZBP1), a molecule already known to play a regulatory role in how the immune system responds to influenza. Specifically, ZBP1detects influenza viruseswithin the lungs and signals infected epithelial and immune cells to self-destruct. This induced cell death eliminates the virus and promotes recruitment of additional immune cells to the infection site.

Building off this finding that ZBP1 is important for fighting viral infection, we found that macrophages infected withYersinia pseudotuberculosis, a type of bacteria that causes foodborne illness, also use this protein to initiatecell death. This limits bacterial replication while also sendinginflammatory signalsthat help clear bacteria.

These findings raise the possibility that ZBP1 may play a dual role in how the body responds to viral and bacterial infections. Its possible that treatments that increase ZBP1 in certain types of cells may be useful in managing bacterial and viral coinfections.

Hayley Muendleinis an assistant research professor of immunology in the Tufts University Graduate School of Biomedical Sciences.

This article was originally published onThe Conversation. Read theoriginal story.

More:
COVID-19 and Flu Viruses Often Have a Deadly Accomplice: Bacterial Infections - Tufts Now

Read More...

Eat This Fruit Daily to Stay Sharp and Live Longer, Research Find – The Beet

Friday, August 19th, 2022

Eating grapes is potentially the cheapest, easiest way to improve your immune system, your metabolism, and your brain health. Foraround $2.09 per pound, grapes contain a bounty of essential nutrients including Vitamin C, antioxidants, calcium, and more. Now, three new studies suggest that just adding two cups of grapes to a high-fat diet can provideremarkable health benefits, surpassingour understanding of the benefits of grapes.

Dr. John Pezzuto examined the health benefits of grapes with a team of researchers from Western New England University. The three studies focus on lifespan, metabolism, fatty liver disease, and brain health, revealing that grape consumption yielded reductions in fatty liver and extended lifespans. To conduct the studies, the researchers analyzed how grape consumption altered gene expression in mice. Despite not conducting human tests, the researchers emphasize that these resultscan reliablytranslate to human health issues.

We have all heard the saying you are what you eat,' which is obviously true since we all start out as a fetus and end up being an adult by eating food, Western New England University Researcher and senior author of three new studies Dr. John Pezzuto said. But these studies add an entirely new dimension to that old saying. Not only is food converted to our body parts, but as shown by our work with dietary grapes, it actually changes our genetic expression. That is truly remarkable.

Pezzutos first study concluded that grape consumption triggered unique gene expressions in the mice. This study found that grape consumption led to a reduced risk of fatty liver disease and expanded the overall lifespan of the animal consuming the grapes. To properly conduct the study, the animals followed a high-fat western style diet. Published in Foods, this study claims that grape consumption can modulate the adverse effects of a traditional Western diet, preventing oxidative damage.

What is the effect of this alteration of gene expression? Fatty liver, which affects around 25% of the worlds population and can eventually lead to untoward effects, including liver cancer, is prevented or delayed, the researchers stated. The genes responsible for the development of fatty liver were altered in a beneficial way by feeding grapes.

The second study, published in Food & Function, found that the consumption of grapes changes metabolism. When Pezzuto and his research team introduced grapes to mice following high-fat diets,researchers found increased levels of antioxidant genes in the mice. The study concluded that grapes help reprogram the metabolism of the gut microbiota, increasing the efficiency of the liver and energy production.

Many people think about taking dietary supplements that boast high antioxidant activity, Pezzuto said. In actual fact, though, you cannot consume enough of an antioxidant to make a big difference. But if you change the level of antioxidant gene expression, as we observed with grapes added to the diet, the result is a catalytic response that can make a real difference.

Published in the journal Antioxidants, the final study observed how grape consumptionbenefits brain function. Theresearch highlights that a high-fat diet presents negative behavioral and cognitive pressures on the brain. In contrast, grape consumptionhelps alleviate these pressures, havinga positive effect on the brain and brain metabolism. The researchers noted that this initial conclusion will require more research to determine the extent of the positive impacts.

Although it is not an exact science to translate years of lifespan from a mouse to a human, our best estimate is the change observed in the study would correspond to an additional 4-5 years in the life of a human, Pezzuto said. Precisely how all of this relates to humans remains to be seen, but it is clear that the addition of grapes to the diet changes gene expression in more than the liver.

This February, a study found that a mostly plant-based diet can prolong life expectancy by over 10 years. The team of Norweigan researchers found that introducing more plant-based foods earlier in life helps cut down the risk of life-threatening disease and improves your overall health. Following an "optimal" diet defined as primarily plant-based a little fish showed long-term health benefits, whereas diets high in red or processed meat showed an inverse relationship.

Another study from last March found that eating more plant-based is key tomaintaining a healthy gut. This study concluded that by improving gut health, you can improve longevity and prolong your lifespan. The researchers claim that building a healthy microbiome at an earlier age is essential to better health in old age.

For more plant-based happenings, visit The Beet's News articles.

Here are the best foods to eat on repeat, to boost immunity and fight inflammation. And stay off the red meat.

See the original post:
Eat This Fruit Daily to Stay Sharp and Live Longer, Research Find - The Beet

Read More...

The Next COVID-19 Booster Shots Will Target Omicron: What to Know – Healthline

Friday, August 19th, 2022

With the countrys third COVID fall approaching, the United States is expected to soon start ramping up its autumn COVID-19 booster campaign.

This years rollout will include something new. Moderna and Pfizer-BioNtech are working on bivalent boosters that include both the original vaccine formula and a component that targets the Omicron BA.4 and BA.5 subvariants of the coronavirus.

While the Biden administration has yet to reveal details of the rollout plan (expect more on that later), heres what we know so far.

The Omicron variant has overcome much of the protection against infection offered by two doses of the mRNA vaccines (such as Moderna and Pfizer-BioNTech).

A first booster restores some of that protection, but this wanes considerably within about three months after vaccination.

In spite of that, Dr. David Cutler, a family medicine physician at Providence Saint Johns Health Center in Santa Monica, Calif., told Healthline that the current vaccines continue to offer strong protection against severe illness and death.

This is especially true of the boosters.

In May 2022, unvaccinated people were six times more likely to die of COVID-19, compared to people vaccinated with at least a primary series (for most, two doses of the mRNA vaccines), according to the Centers for Disease Control and Prevention (CDC).

Among people 50 years and older, unvaccinated people were 29 times more likely to die of COVID-19 than those who had received the primary series and at least two booster doses, agency data showed.

The current COVID-19 vaccines and boosters are based on the original strain of the virus. As CDC data shows, these still offer strong protection against severe disease caused by Omicron.

However, in order to better target the variants likely to be circulating in the fall, the Food and Drug Administration (FDA) asked vaccine makers in June 2022 to update their boosters to include a component that targets the currently circulating Omicron BA.4 and BA.5 subvariants.

We think the Omicron-specific boosters will improve immunity against the existing Omicron variants. This may be particularly helpful during the anticipated winter surge, Dr. Jimmy Johannes, a pulmonologist and critical care medicine specialist at MemorialCare Long Beach Medical Center in California, told Healthline.

However, not all scientists agree that Omicron-specific boosters will provide greater protection than the current ones.

Cutler thinks the strongest benefit of Omicron-specific boosters will be for people who are unvaccinated or have not received the full primary series and any booster(s) for which they are eligible.

One problem with choosing which booster to use in the fall is its impossible to know for certain which variants will be circulating by then, although some experts expect it to be a descendant of one of the currently circulating Omicron variants.

Data presented at an FDA vaccine advisory committee meeting in June 2022, though, suggests that vaccination with a variant-specific booster such as one targeting Omicron might lead to a broadened antibody response against the coronavirus.

Data from Moderna shows the potential for this kind of broader immune response. The companys bivalent Omicron BA.1 booster also produced a higher level of neutralizing antibodies against BA.4 and BA.5 than the original booster, according to preliminary data.

On August 15, the United Kingdoms Medicines and Healthcare Products Regulatory Agency approved Modernas bivalent Omicron BA.1 booster for use in adults.

The Moderna and Pfizer-BioNTech boosters based on the original strain of the coronavirus are currently available for anyone who is eligible for a first or second booster.

The bivalent boosters from those companies are expected to be available in early to mid-September, Dr. Ashish Jha, White House COVID-19 response team coordinator, said in a virtual discussion with the U.S. Chamber of Commerce Foundation on August 16.

Before these boosters can be rolled out, though, the FDA will need to authorize them and the CDC will need to sign off on their use.

The fourth vaccine in the country, Novavaxs protein-based vaccine, was authorized by the FDA on July 13, 2022 for use as a two-dose primary series. This vaccine is based on the original strain of the coronavirus.

The company announced the next month that it had applied for FDA authorization of this vaccine as a booster. It is also testing an Omicron-specific vaccine and a bivalent vaccine that targets Omicron and the original strain, the company said in a release.

Everyone currently eligible for a COVID-19 booster will still be eligible in the fall, including:

Johannes said anyone at risk of severe COVID-19, or complications of a coronavirus infection, should consider getting boosted when the bivalent vaccine is available.

This includes older adults, as well as those with chronic medical conditions such as heart disease, liver or kidney disease, a chronic respiratory condition, cancer, an immune compromising condition, high blood pressure or diabetes.

The Biden administration is also expected to open up second boosters this fall to adults under age 50 when the bivalent vaccines are available. This expansion of eligibility was put on hold when vaccine makers said they could deliver the bivalent vaccines in early fall.

When the bivalent boosters are available in the fall, these will be used for all booster shots in the United States, including first and second boosters.

Pregnant women are also eligible for boosters.

The [COVID-19 mRNA] vaccines have now been given to tens of millions of pregnant women. They are extremely safe, said Jha during the online Chamber of Commerce call. We have seen little to no side effects [in pregnant women], the same side effects that most of us get the sore arm, sometimes 24 hours of feeling fatigued or a little bit run down.

Bolstering the safety profile of these vaccines, a large study from Canada published August 17, 2022, in The BMJ found that women who received a COVID-19 vaccine during pregnancy did not have a higher risk of having a preterm birth, a baby who was small for their gestational age at birth, or a stillbirth.

No details are available yet on the fall booster rollout, but it will likely be similar to the initial booster release last year, with vaccines mainly available at doctors offices and pharmacies. Some mass vaccinations may also happen in certain locations.

To find a vaccination site near you, check out the federal Vaccines.gov or your states COVID-19 vaccine website.

Its difficult to know what the coronavirus will do in the fall will there be a large spike early in September or will a new variant emerge? In addition, theres no guarantee that the bivalent vaccines will definitely be available in September.

As a result, the CDC recommends getting boosted as soon as you are eligible, with whichever booster is available. This is especially important for adults 50 years and older or those with compromised immune systems.

It can take one to two weeks after receiving a booster for your immune system to be fully primed. So if you are eligible now and get boosted, you will be better protected should cases surge as we head into the fall and winter.

You can always get the bivalent vaccine when it is available. Jha said you will want to space out those two boosters at least a little bit, probably 4 to 8 weeks.

The CDC may also weigh in on the timing between boosters when it reviews the data on the Omicron-specific boosters.

Jha said if you are planning on getting the seasonal flu shot this fall, you can definitely get this alongside a COVID-19 booster.

The CDC recommends that people get vaccinated against the flu by the end of October to ensure they have strong immune protection at the peak of the flu season, which generally happens in February.

Scientists dont know yet if the coronavirus that causes COVID-19 will follow a similar seasonal pattern, but cases have tended to increase in colder parts of the country as people head indoors for the fall and winter.

Although the FDA asked vaccine makers to update their boosters to include an Omicron-specific component, it did not advise them yet to update the vaccine for the primary series.

This suggests that unvaccinated people will receive the original vaccine, which the agency said provides a base of protection against serious outcomes of COVID-19.

Read more here:
The Next COVID-19 Booster Shots Will Target Omicron: What to Know - Healthline

Read More...

What to eat when you have COVID – Medical News Today

Friday, August 19th, 2022

Eating a nutrient-dense diet can help someone recover from COVID-19 by supporting their immune system and managing inflammation. This may be particularly important if they lose their sense of taste or smell and have the temptation to eat stronger tasting, less nutritious foods.

People can support their bodies in recovery at home by eating a nutritious diet. This article looks at what experts advise to eat when individuals have COVID-19.

This article also details the symptoms of COVID-19, how the loss of taste and smell affects diet, what to do if vomiting occurs, and some frequently asked questions.

Eating a nutritious diet is an essential consideration in recovering from COVID-19. Research suggests that insufficient nutrition is a risk factor for severe COVID-19, and key nutrients can help support the immune system and manage inflammation.

The following foods and nutrients may help a person recover from the disease.

According to a 2022 review, experts associate a diet involving healthy, plant-based foods with a lower risk and severity of COVID-19. In addition, the study found that a healthy, plant-based diet may particularly benefit people with higher socioeconomic deprivation.

A diet high in saturated fat increases angiotensin-converting enzyme, the main entry point for coronavirus into cells. Research suggests that diets, such as the Mediterranean diet, which are low in saturated fat and high in nutrients from plant foods, can provide more antioxidants for the body. Antioxidants may help fight viruses and support the immune system.

Therefore, eating the following foods can provide fiber, essential vitamins and minerals, and phytochemicals, which are helpful compounds that plants produce.

Learn more about the Mediterranean diet.

According to a 2020 review, high quality proteins, such as fish, eggs, and lean meat, are an essential part of an anti-inflammatory diet that helps produce antibodies and fight off infection.

Learn more about tips and tricks to eat more protein.

The review also notes that dietary fiber consumption correlates with lower mortality from infectious and respiratory diseases. Consuming fiber can also lead to more favorable gut bacteria, which lowers inflammation. People can consume beneficial fiber in:

Learn more about high fiber foods.

Omega-3 fatty acids, especially eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), may have beneficial effects on COVID-19. These include:

Oily fish is a high source of omega-3 fatty acids. Additionally, plant-based foods, such as flaxseed, walnuts, and hemp, contain alpha-linolenic acid, which the body can convert to EPA and DHA. People can also take an omega-3 supplement from fish or algae.

Learn more about omega-3-rich foods.

Some studies have found that vitamin C can shorten the duration of colds and may improve respiratory symptoms. Vitamin C acts as an antioxidant and regulates the immune system and gene expression.

Other research suggests that vitamin C may help improve inflammation markers in people with coronavirus, but people should not consider it as a treatment in supplement form.

However, people can include vitamin C-rich foods in their diet when they have COVID-19 to help support their immune system. These include:

Learn more about foods high in vitamin C.

Vitamin D plays an important role in the bodys immune system. Scientists previously suggested that dietary sources of vitamin D were particularly important during the pandemic because many people had less exposure to the sun during the lockdown.

People get vitamin D from exposure to the sun and foods such as beef liver, egg yolks, cheese, and fortified breakfast cereals.

Learn more about foods high in vitamin D and other sources.

Some research suggests zinc may reduce viral replication and gastrointestinal and respiratory symptoms of COVID-19. However, most research has focused on zinc supplements, so it is difficult to say whether eating zinc foods can help.

However, zinc is an essential mineral for immune function, and eating foods that contain the substance may support recovery from illness.

Foods containing zinc include:

Learn more about foods high in zinc.

A 2020 meta-analysis of research estimates that 48% of patients with COVID-19 globally experienced a loss of smell and around 41% experienced a loss of taste. In some people, these symptoms may persist as part of long COVID.

Learn more about how COVID causes loss of taste and smell.

People who experience a loss of smell or taste may go off foods they usually eat or prefer foods with more salt, sugar, or fat, as they may be able to taste these more. However, individuals need to ensure they eat plenty of fruit and vegetables for their vitamin and antioxidant benefits. Avoiding too many high sugar or high fat foods is also advisable, as these can be inflammatory.

Taste disorder experts advise eating fruits and vegetables individually rather than as combination dishes such as casseroles and one-pots. This is because combination dishes hide individual flavors and dilute the taste.

Additionally, people can try adding more robust flavors such as citrus, herbs, and spices.

Some people experience nausea or vomiting as symptoms of COVID-19. They may not feel like eating but should ensure they hydrate with fluids.

Individuals should consult a doctor if they have excessive vomiting or go off food for longer than expected.

Learn more about food poisoning versus COVID-19.

Here are some frequently asked questions about COVID-19 and food.

According to the CDC, most people with mild to moderate COVID-19 can spread SARS-CoV-2, the virus that causes the disease, no more than 10 days after symptom onset.

However, most individuals with more severe to critical illnesses are likely to be able to spread the virus no more than 20 days after their symptoms began.

The Food and Drug Administration (FDA) advises that there is no evidence of food packaging having associations with the transmission of SARS-CoV-2.

However, people can wipe down food packaging with an antibacterial wipe as an extra precaution.

Research suggests optimal nutrition and dietary nutrient intake can affect the immune system. So eating a nutritious, balanced diet can help people fight the virus.

When someone has COVID-19, they should aim to eat a nutritious, balanced diet that supports their immune system.

Nutrients, such as vitamin C, zinc, and omega-3 fatty acids, can help the process of recovery. Similarly, a person should ensure adequate fiber and protein from nutritious sources.

If someone has lost their sense of taste or smell, they should avoid eating too many inflammatory foods such as sugar and fat. Instead, they may want to try eating individual fruits and vegetables rather than one-pot meals that disguise their flavor.

Read more here:
What to eat when you have COVID - Medical News Today

Read More...

3 Ways Good Inflammation Helps Your Body – First For Women

Friday, August 19th, 2022

These days, the word inflammation is enough to make anyone nervous. A growing body of research associates it with diabetes, heart disease, rheumatoid arthritis, asthma, and more leading us to buy anti-inflammatory supplements and eat antioxidant-rich foods. Many of these supplements and eating habits are healthy, but theres an important point that were missing: Good inflammation also exists, and its very important for our bodies. Heres why.

As explained in a 2018 Oncotarget research paper, inflammation is a biological response of the immune system. And it gets useful when an infection enters the body.

Certain signaling molecules in the immune system will recognize foreign molecules in the body as pathogens. When this happens, the molecules issue an alarm to innate immune cells (which recognize certain molecules on many different pathogens think of them as general fighters), which begin attacking the infection. This response results in inflammation, which takes the form of fever, chills and sweats, sinus congestion, and other symptoms.

While innate immune cells do a good job of attacking infections, they arent the best equipped for targeting specific pathogens. Thats where T and B cells come in. (Think of these as specialized fighters). T cells either target specific pathogens and infected cells or help control the immune response. B cells create customized antibodies (proteins) that attach to pathogens in order to help destroy them. T and B cells take longer to respond to an infection, but are crucial to clearing it out of the body.

In addition, the immune system can recognize toxic compounds that enter the body or sit on top of the skin. This recognition process causes an inflammatory response (such as swelling or redness). With time, specific cells help clear the toxic molecules to prevent them from causing further damage to the body.

If youve ever scratched your leg or twisted your ankle, youre familiar with the inflammation that follows. The body sends blood, fluid, and white blood cells to the injured area to start mitigating the damage and begin repairs.

While pain, redness, and swelling arent pleasant, theyre an important step in the healing process. Without an inflammatory response, our cuts and bruises would never heal.

The key difference between good and bad inflammation appears when an inflammatory response becomes chronic. Acute inflammation is generally good, because its a useful response to immediate injuries or infections. Chronic inflammation, however, occurs when the inflammatory response goes on for too long, or the response is too great.

Examples of this include allergic reactions, rheumatoid arthritis, and heart disease. During an allergic reaction, the immune system inaccurately detects an allergen (like peanut butter) as a foreign invader and mounts a response so huge that it harms the body. In heart disease, sustained, low levels of inflammation can irritate blood vessels and promote the formation of plaque. And in rheumatoid arthritis, the immune system attacks the lining of the joints.

The takeaway? Finding ways to reduce chronic inflammation is a good thing. Just remember that good inflammation happens for a reason.

View post:
3 Ways Good Inflammation Helps Your Body - First For Women

Read More...

Bioinformatics analyses of potential ACLF biological mechanisms and identification of immune-related hub genes and vital miRNAs | Scientific Reports -…

Friday, August 19th, 2022

ACLF is a systemic inflammatory disease accompanied by immune dysfunction and disturbances in energy metabolism. ACLF has a high short-term mortality, which increases with the incidence of failing organs. Although many studies regarding ACLF have been performed, its underlying mechanisms remain to be fully explored. Meanwhile, it has been demonstrated that a strong immune response is a key mechanism of ACLF18,19. Therefore, we explored the intrinsic mechanisms of immune cell infiltration in ACLF using an effective informational biology approach.

Herein, we identified macrophage-associated co-expressed gene modules in ACLF for the first time using a combination of WGCNA and CIBERSORT. We identified immune-related key genes and provided new pathways for future studies on effective targets for ACLF treatments. After bioinformatics and qRT-PCR experiments, 10 immune-related hub genes were identified and mir-16-5p and mir-26a-5p were validated. Altogether, these results might provide new strategies for understanding the pathogenesis of ACLF and developing targeted therapeutic molecules.

In the present study, we evaluated potential pathways and biological processes of ACLF using enrichment analyses. GSEA is characterized by the analysis of collections of genes rather than individual genes, which helps to avoid the inability to reproduce individual high-scoring genes due to poor annotation. In the GSE142255 dataset, the GSEA indicated that immune response, inflammatory pathways, and metabolic pathways were mainly involved in ACLF. Then, we found that the downregulated DEGs were mainly engaged in immune response and inflammatory reaction, while upregulated ones regulated biosynthetic and substance metabolism pathways. These results reflected two major biological processes that co-occurred during the progression of ACLF regulated by different genes: imbalance of immune-inflammatory response and energy metabolism. According to the BP analysis, immune cell activation, differentiation, proliferation, and migration were the major biological processes in ACLF, leading to an expanding inflammatory response. Recently, it was reported that excessive activation of the immune response not only causes a systemic inflammatory response, which subsequently mediates immune-related tissue damage, but also leads to high energy demand. Consequently, the immune system competes with peripheral organs for energy, triggering an immune-related energy crisis in the organism and increasing the risk of organ failure18,20,21. Overall, it was suggested that the hyperimmune response and dysfunctional energy metabolism in ACLF are biologically coupled processes, largely influencing ACLF progress.

CIBERSORT is a widely used deconvolution machine algorithm for estimating the composition of immune cells. It shows superior performance in the identification and fine delineation of immune cells when processing highly noisy mixture data8,22. Here, the CIBERSORT results showed that the population of M0 and M1 macrophages was significantly increased in ACLF patients compared to healthy subjects. We also labeled markers on the surface of macrophages by immunofluorescence and validated their increase in M1 macrophages in the liver of ACLF rats. Macrophages can be polarized into M1 or M2 phenotypes. M1 macrophages can release significant influxes of inflammatory factors and induce cytokine storms with pro-inflammatory effects. On the other hand, M2 macrophages secrete tissue repair factors and exhibit anti-inflammatory and reparative properties23,24. Kupffer cells, a type of macrophage that resides in the hepatic sinusoids, mainly perform innate immune and inflammatory responses25. To search for highly related gene modules, WGCNA identifies similar gene clusters and gene modules by hierarchical clustering. WGCNA also supports the analysis of correlations between gene modules and phenotypic traits26,27. To identify gene clusters associated with macrophages, we performed WGCNA and identified gene modules closely related to M1 macrophage polarization, including the coral1 (containing 3631 genes) and darkseagreen4 (containing 307 genes) modules. Based on the WGCNAs gene modules, we screened immune-related DEGs and constructed a PPI network to find ACLF immune-related hub genes.

Ten hub genes were screened using CytoHubba: RSL1D1, RPS5, CCL5, HSPA8, PRKCQ, MMP9, ITGAM, LCK, IL7R, and HP (Table 3). The differential expression of hub genes was further confirmed by qRT-PCR in ACLF rats. Overall, MMP9, ITGAM, and IL7R were highly expressed during ACLF. Furthermore, ACLF has high 28-day mortality that is closely related to the degree of organ failure in patients. Hence, we used the GSE168048 microarray containing gene expression data of ACLF patients who survived or died at 28days for further investigation. We verified that the expression of RPS5, PRKCQ, MMP9, LCK, ITGAM, IL7R, and CCL5 differed between surviving and deceased patients, suggesting that these genes might be closely related to ACLF progression and could be used to predict ACLF survival status at 28days. Notably, downregulated genes were mostly involved in the promotion of immune response, while the upregulated gene, MMP9, was associated with hepatocyte necrosis. These results suggested that the coexistence of immune paralysis and cell necrosis is a potential ACLF mechanism leading to poor prognosis.

Moreover, miRNAs are potential targets in numerous diseases and control various biological processes. As short-chain RNAs with a coding length of only about 22 nucleotides, miRNAs cannot directly be translated into proteins, but rather regulate protein synthesis by disrupting the stability of target mRNAs and inhibiting their translation through complementary pairing28. Studies have explored the relationship between miRNAs and diseases and proposed the use of miRNAs as a biomarker for disease diagnosis and prognosis as well as a small molecule drug target29. Considering the time and cost of experimental studies, we adopted a database approach combined with experimental validation to study miRNAs that were significantly altered in ACLF. The miRNet 2.0 integrates data from 15 prediction databases and provides visual analytics to enable a more comprehensive and convenient evaluation of the interactions between miRNAs, mRNAs, lncRNAs, and transcription factors15. Herein, we used miRNet 2.0 to construct a miRNA-hub genes network to explore potential miRNAs related to ACLF. During the validation, two miRNAs were significantly altered in ACLF rats: mir-16-5p presented increased expression and mir-26a-5p showed decreased expression. M1 macrophages can transfer mir-16-5p to gastric cancer (GC) cells by secreting exosomes and triggering a T-cell immune response to suppress tumor formation by decreasing the expression of PD-L130. It has been demonstrated that mir-26a-5p decreases with ACLF progression and is associated with worsening liver function and increasing liver disease severity31. However, further studies are needed to validate the potential association between miRNA regulatory networks and ACLF.

Predicting potential disease-associated miRNAs is very meaningful and challenging. Thus, researchers have developed several computational methods and models to perform those predictions. These models can be classified into four categories: score functions, complex network algorithms, machine learning, and multiple biological information29. For example, Chen et al.32 proposed an inductive matrix filling model (IMCMDA) for miRNA-disease association prediction. By integrating miRNA and disease similarity information into the matrix-populated objective function, a low-dimensional representation matrix of miRNAs and diseases was obtained, which was finally combined into a miRNA-disease association score matrix. Chen et al.33 improved the HGIMDA model and further provided the MDHGI model. This model first decomposes the miRNA-disease association matrix to remove data noise, then uses the topological information implied to make predictions through heterogeneous graph inference. It combines machine learning with network analysis methods to make effective predictions for new disease-miRNA associations. Further, Chen et al. proposed an Ensemble of Decision Tree-based MiRNA-Disease Association prediction (EDTMDA) model34 based on the construction of multiple decision trees by randomly selecting negative samples, miRNA features, and disease features, and by dimensionality reduction of the features. The mean of the predicted values from these decision trees is used as the miRNA-disease association score. This model incorporates feature dimensionality reduction into integrated learning to remove noise and redundant information in the learning process and reduce the computational complexity of the model with higher prediction accuracy. Moreover, Liu et al.35 proposed a DFELMDA-based deep forest integrated learning approach to infer miRNA-disease correlations. This model trains a random forest by constructing two auto-encoders based on miRNAs and diseases, extracting low-dimensional feature representation, and finally predicting potential miRNA-disease associations through the random forest. This model combines feature and deep forest-integrated learning models to enhance the prediction accuracy. Bioinformatics-based prediction methods are constantly evolving. Nevertheless, different models have almost different predictive performance for the same datasets. Hence, it is not only necessary to collect large-scale experimental data but also consider other algorithms to improve predictive performance for specific diseases.

Besides the methods covered in this study, the multi-field predictive research of bioinformatics offers a unique perspective on the exploration of diagnostic and therapeutic tools for diseases, not only for ACLF. Currently, with the development of genome-wide technologies, there is an increasing need to explore models that detail the exact mechanisms in which genes and proteins interact to form complex living systems. A gene regulatory network (GRN) is a network of interactions between gene molecules. An improved Markov blanket discovery algorithm based on IMBDANET has been proposed and can effectively distinguish between direct and indirect regulatory genes from GN and reduce the false-positive rate in the network inference process36. Additionally, RWRNET is an algorithm of Random Walk with Restart (RWR) modified by restart probability, initial probability vector, and roaming network applied to GRN that continuously maps the global topology of the network and estimates the affinity between nodes in the network through circular iterations until all nodes are traversed37. In contrast, IMBDANET uses a Markov blanket discovery algorithm for network topology analysis and processing, identifying direct and indirect regulatory genes while solving the problem of isolated nodes. On the other hand, RWRNET focuses on global network topology information but it cannot handle isolated nodes. Finally, the integration of different methods can be more beneficial for the prediction of gene regulatory relationships.

Here, we combined WGCNA and CIBERSORT algorithms and employed GSEA, KEGG, and GO enrichment analyses to explore immune-related hub genes and potential biological mechanisms in ACLF. The hub genes and miRNAs involved in ACLF regulation were also further validated. Since there are few studies regarding ACLF mechanisms, adopting bioinformatics analyses provided valid information and guidance for our research. However, our current study also has some limitations. First, we used an animal model rather than samples from humans to validate the ACLF immune-related hub genes, and the results from animal studies should be treated with caution. Furthermore, although these hub genes and miRNAs were altered and might be involved in the development of ACLF, whether these genes can be new therapeutic targets for ACLF still needs to be explored. Therefore, further experiments are required to validate our findings and explore potential ACLF mechanisms.

Original post:
Bioinformatics analyses of potential ACLF biological mechanisms and identification of immune-related hub genes and vital miRNAs | Scientific Reports -...

Read More...

Nasal vaccines could snuff out COVID, but the hurdles are not to be sneezed at – Sydney Morning Herald

Friday, August 19th, 2022

The largest army of immune cells in the body work together to filter out all the pathogens we inhale every day, defeating viruses and other invaders without us ever knowing.

But viruses have evolved tricks of their own, allowing them to infect the cells and use its molecular machinery to copy itself over and over before spreading down into the lungs and the rest of the body.

Current generation COVID-19 vaccines offer fantastic protection against serious illness and death by recruiting antibodies in the blood that can block the virus spreading to the organs. But these vaccines struggle to generate antibodies in the nose.

The big issue is infection. The current gen of vaccines dont stop infection, said Associate Professor Nathan Bartlett, head of viral immunology at Hunter Medical Research Institute and the University of Newcastle. The virus can continue to circulate, attacking the vulnerable and finding new ways to mutate around our defences.

At least eight nasal vaccines for COVID-19 are in development, according to the World Health Organisation.

They include a phase 1 trial of Tetherexs nasal COVID-19 vaccine SC-Ad6-1 led by the University of Queenslands Associate Professor Paul Griffin. The spray contains a harmless virus modified to look like SARS-CoV-2. The virus infects nose cells and replicates theoretically prompting the immune system into a strong response.

People wear face masks in Melbourne in July.Credit:Getty

Bartlett is working with ENA Respiratory to develop INNA-051, a nose spray full of molecules that bind to cell receptors that trigger the bodys powerful innate immune system, which is capable of fighting off viruses without needing antibodies or T cells. It gives the immune system a head start, Bartlett said.

However, that head start lasts only a week. Bartlett expects the spray would need to be used every week to maintain protection.

In animal trials, the treatment dramatically reduced COVID-19s ability to replicate in the nose. The treatment Bartlett is careful not to call it a vaccine is now in phase 2 clinical trials in humans.

Nasal vaccines face a key challenge: getting a strong and long-lasting immune response in the nose.

The nose is constantly exposed to viruses, bacteria and pollution, so every time we breathe in, immune cells there are much less aggressive than in other parts of the body.

All the stuff were breathing in, if we responded aggressively, wed have a lot more allergies, Griffin said. And antibodies generated in the nose are typically short-lived.

Another problem is getting the dose right. This is easy with a syringe into a vein, but very difficult when spraying fluid into the nose. What if the users nose is blocked? What if they sneeze? The vaccines also need to be carefully designed to not cause an allergic reaction.

And then there is the mucus itself. Vaccines need to penetrate it to get an immune response and then must stay around long enough to really fire up the system a tough task when mucus is constantly being cleaned out of the nose.

Loading

Griffin said he was confident well get at least one intranasal protection soon. Bartlett is less so. He points out that regulators are unlikely to offer quick emergency approval to new vaccines now and any vaccine that gets approved would likely be out of date immediately, as the virus continues to mutate.

Whether it will have a huge impact on this pandemic, Im not sure, he said. But its critical for protecting us against the next one.

Stay across the most crucial developments related to the pandemic with the Coronavirus Update. Sign up for the weekly newsletter.

Follow this link:
Nasal vaccines could snuff out COVID, but the hurdles are not to be sneezed at - Sydney Morning Herald

Read More...

Paxlovid Rebound: RWE Analysis and Alternative Therapies in Development – BioSpace

Friday, August 19th, 2022

Fabian Sommer/Picture Alliance via Getty

Paxlovid is one of a handful of drugs authorized to treat COVID-19 in the United States. As recently experienced by President Biden, its use is not without challenges and sometimes leads to relapse. The President, like many others, tested negative for COVID-19 after five days of Paxlovid treatment, then tested positive again a few days later.

Although such treatment rebound rates are assumed to be low, there is little data to support or dispute that assumption. Researchers at Case Western University looked at the real-world experiences of 13,644 patients who were treated with Paxlovid or Lagevrio (molnupiravir) during the first half of 2022. The study was intended to determine the prevalence of three types of rebound outcomes with these two therapies, each of which has emergency use authorizations as treatments for COVID-19.

Comparing Paxlovid and Lagevrio

In that study (currently in preprinton medRxiv, co-author Pamela Davis, M.D. and colleagues found that the 7-day and 30-day rebound rates associated with Paxlovid treatments, respectively, were 3.53% and 5.4% for COVID-19 reinfection; 2.31% and 5.87% for COVID-19 symptoms; and 0.44% and 0.77% for hospitalizations.

The rebound rates for patients treated with Lagevrio were somewhat higher. The 7-day and 30-day rebound rates for this therapy were 5.86% and 8.59% for COVID-19 infection; 3.75% and 8.21% for COVID-19 symptoms; and 0.84% and 1.39% for hospitalizations. Propensity-score matching rendered these differences negligible, however.

Patients who rebounded from either therapy had a significantly higher prevalence of underlying medical conditions than those without, the authors noted in the paper. However, There is no underlying condition that stands out, Davis told BioSpace. We had thought (the rebound group would be composed of) immunocompromised people, but heart disease, hypertension, mood disorders and so on, all are increased in the rebound group.

The implications upon dosage or duration of treatment have not been determined, she said, but changes should be considered. If I had a patient with risk factors, for whom the disease might be more than a nuisance, I might be tempted to treat for a longer period of time.Also, I would try to start the drug earlier in the course of the disease, when the virus has had less time to replicate, but I have no data to show that would work, she acknowledged.

Davis and her colleagues called for continued surveillance of patients after treatment with either Paxlovid or Lagevrio, and for additional studies to determine the mechanism of action of the rebound as well as the most effective dosing and duration regimen.

Paxlovid is the combination of two drugs: nirmatrelvir, which blocks viral replication, and ritonavir, which slows the degradation of nirmatrelvir. Ian Chan, co-founder and CEO of Abpro, told BioSpace the therapy is like a carpet-bombing approach. It can kill COVID, and healthy cells as well. Because its a small molecule it also can have multiple drug-to-drug interactions, so its high efficacy comes with high toxicity, he noted. There also a risk for COVID to mutate around it, developing resistance as the virus changes.

No one yet knows why Paxlovid rebound occurs but, Our theory is that because this drug is prescribed very early on, there is less chance for the immune system to develop a response (to the virus), Chan said.

One solution to therapeutic rebound, he said, is to develop more types of therapies. COVID-19, unfortunately, is going to be around for a while. We need a battery of different treatments just to be ready for the different possible scenarios.

Abpro is developing monoclonal antibody (mAb) therapies for COVID-19, as well as for immuno-oncology and ophthalmology. The benefit, Chan explained, is that the safety of mAbs has been proven. These are natural molecules your immune system already is producing on a day-to-day basis. Their toxicity generally is very low and efficacy is very high, and they are very targeted therapies, he said. So far, there have been no therapeutic relapses.

Potential Therapies for the Immunocompromised

To treat COVID-19, Abpro is focusing on treatments for the approximately 15 million immunocompromised individuals in the U.S. who, because of their weakened immune systems, dont respond to vaccines.

The companys lead compound, ABP-300, is in Phase II clinical trials. It basically prevents the virus from binding in the body, thus neutralizing it. Other antibodies are undergoing investigational new drug application (IND)-enabling studies.

One of those, ABP-C19-01, is a cocktail of antibodies that lowers patients risk of contracting COVID-19 if they are exposed to the SARS-CoV-2 virus. This therapy isnt a vaccine, Chan stressed. Vaccines are meant to stimulate an immune response, which generates an antibody. This delivers the antibody directly. It is complementary to vaccines.

Another company, 3CL Pharma, a subsidiary of Todos Medical, is developing possible mitigations for Paxlovid rebound that work by supporting the immune system and inhibiting 3CL protease activity. Its solutions include Tollovir and Tollovid.

A Phase II trial of Tollovir, which is being developed to treat hospitalized COVID-19 patients, was completed in May in Israel.

Were developing Tollovir as an alternative for Paxlovid, Gerald Commissiong, CEO of Todos Medical, told BioSpace. It is similar to the nirmatrelvir portion of Paxlovid, but is a botanical, so it works slower. Also, rather than affecting the virus directly, Tollovir works on the 3CL protease, which is responsible for helping a virus that already has replicated to infect other cells.

The other compound, Tollovid, is being developed as an over-the-counter dietary supplement. According to Todos literature, it has very strong 3CL inhibition. It may be taken as a 5-day regimen, or in a daily formulation for immune system support. It may become a rescue agent for Paxlovid rebound.

Continue reading here:
Paxlovid Rebound: RWE Analysis and Alternative Therapies in Development - BioSpace

Read More...

Predicting response to immunotherapy in gastric cancer via multi-dimensional analyses of the tumour immune microenvironment – Nature.com

Friday, August 19th, 2022

Clinico-pathological features of the GC patients

Eighty patients were enrolled in this study between July 2014 and December 2019 (Table1). The median age of the patients was 60 years (range, 5466 years), and most patients were men (76.3%). Among the 60 patients subjected to immunotherapy, 21 were treated with standard-of-care anti-PD-1/PD-L1 antibodies and 39 were treated as part of clinical trials (NCT03472365, NCT03713905). Archived pre-treatment samples were available from all patients. Ten (12.5%) patients were EBV(+) and 11 (13.75%) had confirmed deficient DNA mismatch repair (dMMR) GC.

To investigate the landscape of TIICs within the GC specimens, we quantified the density and spatial location of immune cells in 80 full-face formalin-fixed paraffin-embedded (FFPE) samples via m-IHC staining; the multiplex determination of the sub-cellular expression of 16 proteins was performed (Fig.1a). First, haematoxylin and eosin (H&E)-stained tissue sections were reviewed by two pathologists (S.Y. and H.Y.J.) to identify tumour core (TC), invasion margin (IM), and peri-tumoural normal (N) areas, which we refer to as regions of interest (ROIs) (Fig.1b). The m-IHC panels analysed are depicted in Fig.1cf. A total of 6488 high-power fields (TC: 4477, IM: 993, N: 1018) were imaged for all patients. A supervised image analysis system (inForm) was used to classify each image into tumour nests and stromal areas based on machine learning (Fig.1g). Cell segmentation showed nuclear, cytoplasmic and membranous outlines. Cell phenotyping data were obtained based on the positivity and relative intensity of all markers in one panel. The cell density, calculated for all regions (tumour + stroma), was measured separately in the tumour and stroma. Thereafter, TIICs were analysed at the single-cell level and 26 major populations were characterised (Supplementary Fig.1a).

a Schematic representation of the experimental design and analytical methods used in this study. b Selection of the regions of interest (ROIs) in representative images of haematoxylin and eosin (H&E)-stained formalin-fixed paraffin-embedded tissues. TC, tumour core; IM, invasion margin; N, normal tissue. Scale bar: 3mm. cf Representative composite and single-stained images of the multiplex immunohistochemistry panels used. Scale bar: 200m. g Overview of the automated image analysis pipeline.

To examine the distribution of TIICs within the tumour microenvironment, we analysed their spatial density in the TC, IM, and N areas. The enriched co-occurrence of immune populations defines a structured immune environment (Supplementary Fig.1a). A significant increase in the overall density of CD68+ cells was observed within the TC compared with that in the adjacent normal tissues; an opposite trend was observed for CD8+ and CD20+ cells (Fig.2a). Next, for a higher degree of detail, the distribution of each TIIC was explored. CD8+, CD8+PD-1LAG-3, CD20+ and CD68+CD163+HLA-DR cells accumulated at the IM and decreased toward the TC. In contrast, CD8+PD-1+TIM-3+, CD8+PD-1TIM3+, CD8+PD-1+LAG-3+TIM-3+, CD8+PD-1+LAG-3TIM-3+, CD4+FoxP3+CTLA-4+, CD4+FoxP3CTLA-4+, CD68+, CD68+HLA-DR+CD163 cells accumulated at the TC and decreased toward the IM. Interestingly, a higher density of CD4+FoxP3+ and CD4+FoxP3+PD-L1+ cells was found within the TC than in normal tissues (Fig.2b, Supplementary Fig.1b), highlighting the heterogeneous distribution of TIICs in GC.

a Constitution of the main tumour-infiltrating immune cell (TIIC) populations. KruskalWallis test with the Dunns multiple comparison test. b Density of TIICs across the regions of interest (n=80). TC, tumour core; IM, invasion margin; N, normal tissue. Immunofluorescence staining images refer to the co-expression of the corresponding markers and DAPI (nuclei). Scale bar: 20m. Box and whiskers represent mean1090 percentile. KruskalWallis test with Dunns multiple comparison test. c TIIC density grouped by subtypes. d Overall survival of 80 patients based on the density of TIICs. The individual TIICs were divided into high (>two-thirds of the patients; blue line) or low density (two-thirds of patients; red line). Log-rank (MantelCox) test was used. A two-sided P<0.05 was considered statistically significant.

Additionally, the localisation of TIICs with respect to the tumour nest and stroma areas (defined in Fig.1g) was further examined. CD8+, CD4+ and CD20+ cells were located primarily in the stroma and were less prevalent in the tumour nest. In contrast, CD66b+ cells were more prevalent in the tumour nest than in the stroma (Supplementary Fig.2a).

To evaluate the tumour immune microenvironment in GC, we compared the density of TIICs in the context of distinct clinico-pathological factors (Fig.2c, Supplementary Fig.3ae). Generally, there were few significant differences between Lauren classification, tumour differentiation and tumour location (oesophagogastric junction or not) with respect to densities of TIICs (Supplementary Tables15, Supplementary Fig.4a). Additionally, there were few differences in the density of TIICs between HER2-positive and -negative GC (Fig.2c). Overall, the density of total CD8+, CD4+ and CD68+ cells was associated with the disease stage. Additionally, advanced-stage GC (III-IV) samples showed a higher density of exhausted CD8+ T cells, CD4+FoxP3 cells and so on.

Furthermore, we analysed the density of TIICs in GC of different molecular subtypes (Supplementary Tables68). Interestingly, EBV-positive tumours showed higher densities of CD8+PD-1LAG-3 T cells than EBV-negative ones. EBV (+) GCs were characterised by abundant immune cell infiltration; however, not all EBV (+) patients responded to immunotherapy, indicating that specific immune cell infiltration is needed. Proficient MMR (pMMR) tumours showed a significantly higher abundance of total CD4+, CD68+, CD20+ and CD66b+ cells than dMMR tumours. Higher CD68+ and CD66b+ cells (neutrophils) are known to contribute to resistance to PD-1/PD-L1 treatment in several cancers13,19. We classified patients into four combined positive score (CPS) groups: CPS<1, 1 CPS<5, 5 CPS<10 and CPS10. Remarkably, the abundance of TIICs, including CD8+, CD4+, CD68+, CD20+ and CD66b+ cells, significantly increased with the increase in CPS, indicating a hotter tumour immune environment. However, the comparison between CPS 5-10 and CPS10 did not show a significant difference, providing evidence for the cut-off selection in clinical trials of anti-PD-1/PD-L1-based therapies. Altogether, as shown in Fig.2c, our results suggest that the infiltration pattern of immune cells depends on, but is not restricted to, GC molecular subtypes.

Next, we sought to understand whether the number of TIICs is correlated with patient survival. We found that higher levels of tumour-infiltrating T cell subsets, including CD8+PD-1+LAG-3+TIM-3+, CD4+FoxP3+CTLA-4+ T and CD68+STING+ cells, were associated with inferior overall survival (OS) in 80 patients (Fig.2d, Supplementary Fig.4b). CD8+PD-1+LAG-3+TIM-3+ cells [high vs. low, hazard ratio (HR) 1.98, 95% confidence interval (CI; 1.123.50)] and CD68+STING+ cells [high vs. low, HR 1.83, 95%CI (1.013.33)] were significantly associated with OS, as revealed by multivariate Cox analysis (Supplementary Table9). Collectively, these data highlight the clinical relevance of tumour-infiltrating T cells in the survival of GC patients.

Additionally, we analysed the prognostic value of the density of TIICs in the context of tumour and stromal cells. The data showed a similar trend for CD4+FoxP3CTLA-4+ T and CD4+FoxP3+CTLA-4+ T cells in both contexts. However, higher infiltration of CD8+PD-1+LAG-3+TIM-3+ T cells and CD68+ macrophages was associated with poorer OS with respect to tumour nests. In addition, higher infiltration of CD8+PD-1+TIM-3+ T cells, CD66b+ neutrophils and CD68+STING+ macrophages was related to a shorter OS with respect to the stroma (Supplementary Fig.2b). Therefore, these results highlight the value of studying immune cell density in defined tissue regions.

Given our ability to precisely define the positions of individual tumour cells and TIICs, we next sought to evaluate the clinical significance of the proximity between them. The observation that certain TIICs, including CD68+ cells, were enriched in the tumour region suggested that the proximity of TIICs to tumour cells might influence their phenotype. To further study these localisation patterns, a bioinformatics tool (pdist; see Methods) that determines the nucleus-to-nucleus distances between any two cell types was used. To incorporate both cell proximity and quantity, an effective score parameter was established: the proportion of TIICs near tumour cells (within the defined distance criteria introduced; Fig.3a). In other words, this score was calculated by the number of paired immune cells and tumour cells divided by the total number of immune cells across the whole slides to maintain the spatial variation to a large extent. Therefore, using this formula, a higher effective score indicates that within a certain distance, there is a higher density of tumour cells around the immune cells. Importantly, across the three distances considered (010/020/030m), CD8+PD-1+LAG-3+ T cells and CD66b+ neutrophils were the ones with higher effective scores (Fig.3b).

a Illustration of the distance analysis involving immune and tumour cells. Red dots: tumour cells; green dots: immune cells. The white translucent circle represents the radius. Effective score=number of paired immune cells and tumour cells/number of immune cells. Scale bar: 100m. b The distribution of the effective score of tumour-infiltrating immune cell (TIIC) populations in the tumour core in 10-, 20- and 30m increments (n=80). Error bars represent meanSEM. c Effective score of TIICs in patients grouped by gastric cancer subtypes. EBV, EpsteinBarr virus status; MMR, DNA mismatch repair; CPS, combined positive score. d Overall survival of the 80 patients based on the effective densities (010m and 020m) of TIICs. The individual immune infiltrate values were divided into high (> two-thirds of the patients in the cohort; blue line) or low density ( two-thirds of patients in the cohort; red line). Statistical relevance was defined using the log-rank (MantelCox) test. A two-sided P<0.05 was considered statistically significant.

We also calculated the distance between each TIIC and the closest tumour cell. Neutrophils, B cells and macrophages were located closer to tumour cells. We then analysed the distances between TIICs and tumour cells according to the PD-L1 CPS. In general, TIICs were located closer to tumour cells in patients with CPS10 (compared with the picture with respect to all other groups; Supplementary Fig.6a).

Interestingly, the effective scores also differed between different GC molecular subtypes, including those depending on the EBV, PD-L1 CPS, MMR and HER2 status (Supplementary Figs.5ae, 6b; Supplementary Tables1017). For instance, a significantly higher effective score of exhausted T cells (CD8+PD-1+LAG-3+TIM-3, CD8+PD-1TIM-3+), M1 (CD68+CD163+HLA-DR) and M2 (CD68+HLA-DR+CD163) macrophages within a 20m radius was observed in HER2-negative GC compared with that in HER2-positive GC (Fig.3c, Supplementary Fig.5b).

The combination of multiplexed imaging and machine learning implied that the density of TIICs within GC is linked to patient survival. For further detail, the effective density (the absolute number of TIICs near tumour cells within a 20m radius) was used as an additional measurement. This radius was pre-selected to identify immune cell populations most likely capable of effective, direct, cell-to-cell interactions with tumour cells, consistent with prior studies in multiple gastrointestinal tumour types11,20,21. Curiously, we found that patients with higher effective densities (radius of 020m) of CD68+STING+ macrophages, CD68+HLA-DR+CD163 STING+ macrophages and neutrophils showed significantly shorter OS than those with lower effective densities (Fig.3d). Importantly, the prognostic value was still significant after adjustment using the multivariate Cox model (Supplementary Table18). Other immune cell phenotypes were not associated with OS (Supplementary Figs.6c and 7c). These results indicate that the influence of TIICs on patient survival is dependent not only on the number of TIICs but also on their proximity to tumour cells. Overall, our data highlight that both the location and density of TIICs should be taken into consideration for prognosis predictions.

Human tumours contain exhausted T cells expressing multiple immune checkpoints; it has been proposed that these cells mediate resistance to PD-1 blockade. Thus, next, we investigated whether the density of TIICs and respective effective scores were associated with the clinical outcomes of anti-PD-1/PD-L1 immunotherapy. All 60 patients who received immunotherapy were assigned to the training (n=44, generated retrospectively from 15/11/2016 to 17/7/2019) and validation (n=16, generated prospectively from 29/7/2019 to 19/12/2019) cohorts. Importantly, we ensured that the clinical characteristics of the training and validation cohorts were balanced (Table2). We used logistic regression analysis to assess the association between TIICs and the objective response rate (ORR) in the training cohort. Importantly, we found that the density of CD4+FoxP3PD-L1+ T cells and the effective score of CD8+PD-1+LAG-3 T cells were closely associated with a positive response to anti-PD-1/PD-L1 therapy; conversely, CD8+PD-1LAG-3 T cells and CD68+STING+ macrophages were closely associated with a negative response to anti-PD-1/PD-L1 therapy (Supplementary Table19).

The density of CD4+FoxP3PD-L1+ T cells, CD8+PD-1LAG3 T cells and CD68+STING+ macrophages, and the effective score of CD8+PD-1+LAG3 T cells were used to define a TIIC signature (Fig.4a), with the potential to improve the ability of identifying responders to anti-PD-1/PD-L1 immunotherapy. We used four types of machine learning models and calculated the area under the curve (AUC) of the training and validation cohorts, including extra tree classifier (ETC), AdaBoost classifier (ABC), gradient boosting classifier (GBC) and multi-layer perceptron (MLP) models. In the validation cohort, the average AUCs of the four algorithms were 0.80, 0.85, 0.77 and 0.75, respectively (Fig.4b, c, Supplementary Table20). The corresponding 95% CIs were narrow, suggesting that the TIIC signature can indeed be used to predict the response to immunotherapy (Supplementary Table20). Importantly, the four algorithms showed a similar performance before and after adjusting for the hyper-parameters, indicating the strength of the predictive value of the TIIC signature itself (Supplementary Fig.7a). Furthermore, we explored the predictive power of the TIIC score combined with CPS, EBV status and MMR status. The combined TIIC signature had a better AUC in the ETC, GBC and ABC models, but not in the MLP model (Supplementary Table21).

a Definition of the tumour-infiltrating immune cell (TIIC) signature. Red arrows highlight specific immune cells. b Average area under the curve (AUC) of TIIC signature and combined TIIC signature (TIIC+ EpsteinBarr virus status+mismatch repair status+PD-L1 combined positive score) in the four machine learning models in the validation cohort. c Representative receiver operating characteristic (ROC) curves for the performance of the identified TIIC signature and combined TIIC signature in gastric cancer patients subjected to immunotherapy in the validation cohort. ETC extra tree classifier, GBC gradient boosting classifier, ABC AdaBoost classifier, MLP multi-layer perceptron.

We quantified the contribution of each marker in the prediction models through feature importance using the scikit-learn package (Supplementary Tables22, 23). We outputted the feature importance and the average value of each parameter to present its contribution. As shown in Fig.5a, the effective score of CD8+PD-1+LAG-3 cells had higher feature importance than the density of CD68+STING+, CD4+FoxP3PD-L1+, or CD8+PD-1LAG-3 cells in ETC, GBC and ABC machine learning models. As presented in Fig.5b, the effective score of CD8+PD-1+LAG-3 cells had higher feature importance than that of the other three immune cell types, EBV, MMR and PD-L1 CPS. Thus, the dominant predictive marker is the spatial organisation for response to immunotherapy.

a, b The feature importance of each marker in the prediction model. c, d KaplanMeier curves of the (c) immune-related progression-free survival (irPFS) and (d) immune-related overall survival (irOS) of anti-PD-1/PD-L1-treated patients stratified by the tumour-infiltrating immune cell (TIIC) signature in the validation cohort. Log-rank (MantelCox) test was used for analysis.

We also evaluated the predictive values of other candidate biomarkers. AUCs of 0.58 and 0.76 in the training and validation cohorts, respectively, were defined for PD-L1 CPS (Supplementary Fig.7b). We analysed the treatment response based on EBV status, MMR status and HER2 expression in univariate and multivariable logistic regression models (Supplementary Table24). EBV-positive status and dMMR tended to be associated with a better response. The association of HER2 expression with treatment response was not consistent between univariate and multivariable models. Therefore, taken together, our data suggest that the TIIC signature has a greater power for patient stratification (Supplementary Fig.7b).

Next, we investigated the prognostic use of the TIIC signature; the univariate Cox proportional hazard regression model was used to calculate the HR of each indicator. Then, we used the HR of each indicator as the weight to multiply the value of the indicator itself and then calculated the weighted sum of the four indicators. In this analysis, we categorised patients into high- and low-score groups based on the TIIC signature. The difference in the survival probability over time between the groups was calculated using the KaplanMeier method. As expected, we observed a significant difference in both immune-related progression-free survival (irPFS) and immune-related overall survival (irOS) in the validation cohort (Fig.5c, d, Supplementary Table25). Therefore, the TIIC signature might be useful to identify patients that will show active anti-tumour immune responses a priori.

See the article here:
Predicting response to immunotherapy in gastric cancer via multi-dimensional analyses of the tumour immune microenvironment - Nature.com

Read More...

What Happens When Your Immune System Gets Stressed Out? – Cleveland Clinic

Friday, July 8th, 2022

For mostof us,stressis just apart of life. It can last for a few hours like the time leading up to a final exam or for years like when youre taking care of an ailing loved one.

Cleveland Clinic is a non-profit academic medical center. Advertising on our site helps support our mission. We do not endorse non-Cleveland Clinic products or services.Policy

Stress is sometimesa motivator that helpsyou rise to the occasion. At other times, its simply overwhelming. Whatever the case, if its chronic, it can takea toll on your immune system.

Clinical immunologistLeonard Calabrese, DO, offers insights on how stress impacts your immunity and what you can do to minimize the effect.

Eliminating or modifying these factors in ones life is vital to protect and augment the immune response, he says. Its necessary to buffer the inevitability of the aging process.

Stress occurs when life events surpass your abilities to cope. It causes your body to produce greater levels of the stress hormone cortisol.

In short spurts, cortisolcan boost your immunity by limitinginflammation. But over time, your body can get used to having too much cortisol in your blood. And this opensthe door for more inflammation, Dr. Calabrese says.

In addition, stress decreases the bodys lymphocytes the white blood cells that help fight off infection. The lower your lymphocyte level, the more at risk you are for viruses, including the common cold and cold sores.

High stress levels also can causedepressionand anxiety, again leading to higher levels of inflammation.In the long-term, sustained, high levels of inflammation point to an overworked, over-tired immune system that cant properly protect you.

If you dont control high stress levels, chronic inflammation can accompany it and can contribute to the development and progression of many diseases of the immune system such as:

Under sustained, long-term stress, you also can develop cardiovascular problems, including a fast heart rate andheart disease, as well as gastric ulcers. Youll also be at greater risk fortype 2 diabetes, various cancers and mental decline.

Stress reduction strategies not only give your mind a break, but they can also relieve the pressure on your immune system. You can take steps to reduce short-term and long-term stress, Dr. Calabrese says. Two tactics are most effective:

Stress in acute situations, however, can be healthful and protective, so its not all bad for us. Remember: its chronic stress that we seek to control.

Original post:
What Happens When Your Immune System Gets Stressed Out? - Cleveland Clinic

Read More...

Alcohol and the Immune System – PMC – PubMed Central (PMC)

Friday, July 8th, 2022

Clinicians have long observed an association between excessive alcohol consumption and adverse immune-related health effects such as susceptibility to pneumonia. In recent decades, this association has been expanded to a greater likelihood of acute respiratory stress syndromes (ARDS), sepsis, alcoholic liver disease (ALD), and certain cancers; a higher incidence of postoperative complications; and slower and less complete recovery from infection and physical trauma, including poor wound healing.

This issue of Alcohol Research: Current Reviews (ARCR) summarizes the evidence that alcohol disrupts immune pathways in complex and seemingly paradoxical ways. These disruptions can impair the bodys ability to defend against infection, contribute to organ damage associated with alcohol consumption, and impede recovery from tissue injury. It is our hope that a greater understanding of the specific mechanisms through which alcohol exerts its effects on the immune system may lead to development of interventions to prevent, or at least mitigate, the negative health consequences of alcohol misuse.

Contributors to this issue of ARCR lay the groundwork for understanding the multilayered interactions between alcohol and immune function by presenting an overview of the immune system (see the article by Spiering) and by reviewing current research on the effects of alcohol on innate immunity (see the article by Nagy) and on adaptive immunity (see the article by Pasala and colleagues). As reviewed by Szabo and Saha, alcohols combined effects on both innate and adaptive immunity significantly weaken host defenses, predisposing chronic drinkers to a wide range of health problems, including infections and systemic inflammation. Alcohols widespread effects on immune function also are underscored in the article by Gauthier, which examines how in utero alcohol exposure interferes with the developing immune system in the fetus. This exposure increases a newborns risk of infection and disease; additional evidence suggests that alcohols deleterious effects on immune development last into adulthood.

The gastrointestinal (GI) system is typically the first point of contact for alcohol as it passes through the body and is where alcohol is absorbed into the bloodstream. One of the most significant immediate effects of alcohol is that it affects the structure and integrity of the GI tract. For example, alcohol alters the numbers and relative abundances of microbes in the gut microbiome (see the article by Engen and colleagues), an extensive community of microorganisms in the intestine that aid in normal gut function. These organisms affect the maturation and function of the immune system. Alcohol disrupts communication between these organisms and the intestinal immune system. Alcohol consumption also damages epithelial cells, T cells, and neutrophils in the GI system, disrupting gut barrier function and facilitating leakage of microbes into the circulation (see the article by Hammer and colleagues).

These disruptions to the composition of the gut microbiota and to gut barrier function have important implications beyond the intestinal system. For example, Nagy discusses how the leakage of bacterial products from the gut activate the innate immune system in the liver, triggering inflammation that underlies ALD, a condition that affects more than 2 million Americans and which eventually may lead to liver cirrhosis and liver cancer. Infection with viral hepatitis accelerates the progression of ALD, and end-stage liver disease from viral hepatitis, together with ALD, is the main reason for liver transplantations in the United States. The article by Dolganiuc in this issue explores the synergistic effects of alcohol and hepatitis viruses on the progression of liver disease as well as alcohol consumptions injurious effect on liver antiviral immunity. Mandrekar and Ju contribute an article that homes in on the role of macrophages in ALD development, including recent insights into the origin, heterogeneity, and plasticity of macrophages in liver disease and the signaling mediators involved in their activation and accumulation.

In addition to pneumonia, alcohol consumption has been linked to pulmonary diseases, including tuberculosis, respiratory syncytial virus, and ARDS. Alcohol disrupts ciliary function in the upper airways, impairs the function of immune cells (i.e., alveolar macrophages and neutrophils), and weakens the barrier function of the epithelia in the lower airways (see the article by Simet and Sisson). Often, the alcohol-provoked lung damage goes undetected until a second insult, such as a respiratory infection, leads to more severe lung diseases than those seen in nondrinkers.

In a clinical case study reviewed in this issue, Trevejo-Nunez and colleagues report on systemic and organ-specific immune pathologies often seen in chronic drinkers. In such patients, alcohol impairs mucosal immunity in the gut and lower respiratory system. This impairment can lead to sepsis and pneumonia and also increases the incidence and extent of postoperative complications, including delay in wound closure. HIV/AIDS is a disease in which mucosal immunity already is under attack. Bagby and colleagues review substantial evidence that alcohol further disrupts the immune system, significantly increasing the likelihood of HIV transmission and progression.

Alcoholimmune interactions also may affect the development and progression of certain cancers. Meadows and Zhang discuss specific mechanisms through which alcohol interferes with the bodys immune defense against cancer. They note, too, that a fully functioning immune system is vital to the success of conventional chemotherapy. The clinical management of all of these conditions may be more challenging in individuals who misuse alcohol because of coexisting immune impairment.

Alcohol consumption does not have to be chronic to have negative health consequences. In fact, research shows that acute binge drinking also affects the immune system. There is evidence in a number of physiological systems that binge alcohol intake complicates recovery from physical trauma (see the article by Hammer and colleagues). Molina and colleagues review research showing that alcohol impairs recovery from three types of physical traumaburn, hemorrhagic shock, and traumatic brain injuryby affecting immune homeostasis. Their article also highlights how the combined effect of alcohol and injury causes greater disruption to immune function than either challenge alone.

Not only does the immune system mediate alcohol-related injury and illness, but a growing body of literature also indicates that immune signaling in the brain may contribute to alcohol use disorder. The article by Crews, Sarkar, and colleagues presents evidence that alcohol results in neuroimmune activation. This may increase alcohol consumption and risky decisionmaking and decrease behavioral flexibility, thereby promoting and sustaining high levels of drinking. They also offer evidence that alcohol-induced neuroimmune activation plays a significant role in neural degeneration and that the neuroendocrine system is involved in controlling alcohols effects on peripheral immunity.

Much progress has been made in elucidating the relationship between alcohol consumption and immune function and how this interaction affects human health. Continued advances in this field face several challenges, however. The regulation of immune function is exceedingly complex. Normal immune function hinges on bidirectional communication of immune cells with nonimmune cells at the local level, as well as crosstalk between the brain and the periphery. These different layers of interaction make validation of the mechanisms by which alcohol affects immune function challenging. Significant differences between the immune system of the mousethe primary model organism used in immune studiesand that of humans also complicate the translation of experimental results from these animals to humans. Moreover, the wide-ranging roles of the immune system present significant challenges for designing interventions that target immune pathways without producing undesirable side effects.

By illuminating the key events and mechanisms of alcohol-induced immune activation or suppression, research is yielding deeper insights into alcohols highly variable and sometimes paradoxical influences on immune function. The insights summarized in this issue of ARCR present researchers and clinicians with opportunities to devise new interventions or refine existing ones to target the immune system and better manage alcohol-related diseases.

Read this article:
Alcohol and the Immune System - PMC - PubMed Central (PMC)

Read More...

Hyperactivation of the immune system may cause post-COVID syndromes – EurekAlert

Friday, July 8th, 2022

Investigators at Cedars-Sinai have proposed a theory for howSARS-CoV-2, the virus that causes COVID-19, infects the body. Their hypothesis,published inFrontiers in Immunology, could explain why some people still have symptoms long after the initial infection.

Weve put together different pieces of datato create a bigger picture that may explain what causes some peoples immune systems to go haywire, leading to post-acute syndromes, includingmultisystem inflammatory syndrome in children(MIS-C)and long COVID in children and adults, saidMoshe Arditi, MD,executive vice chair of the Department of Pediatrics for Research,part of Cedars-Sinai Guerin Childrens,and senior author of the paper.

MIS-C is a rare but dangerous condition in children that may occur weeks after infection with SARS-CoV-2.Long COVID-19often referred to as long COVIDis a termused to describe a constellation of health problems that some people experience as a result of their infection withSARS-CoV-2. Symptoms can last months or even years.

SARS-CoV-2 is thought to latch on to cells via spikes that exist on the surface of the virus. These spike proteins are comprised of molecular motifs,stretches of amino acids that make a protein. These tiny molecular motifs may have what the scientists call superantigen characteristics, meaning that the immune system can overreact to their presence.

The spike protein, according to the authors, may also have neurotoxic motifs that can cross the blood-brain barrier and damage brain cells. This hypothesis could explain the brain fog and other neurological symptoms associated with COVID-19 and long COVID.

The hypothesis is based on several published studies on COVID-19 and other diseases caused by viruses. One such study by Arditi and his longtime collaboratorIvet Bahar, PhD,was publishedin theProceedings of the National Academy of Sciencesin 2020. Bahar and Arditi created a computer model showing how molecular motifs onthe spike protein interact with immune cells. The superantigen molecular motifs cause the immune cells to release an abundance ofinfection-fighting proteins known as cytokinesthat fight the virus but alsomay mistakenly attack the bodys organs. In children, this may manifest as MIS-C.

Other studies have reported that people with long COVID may carry fragments of the virus in their gut or other parts of their bodies months after initial infection. Continuous exposure to motifs that lodge themselves in different parts of the body and have superantigen-like properties may cause autoimmune symptoms in people with long COVID and MIS-C, according to the authors.

We need to conduct more research to prove if this is indeed the mechanism that causes long COVID so that we can develop treatments to block it,saidMagali Noval Rivas, PhD,an investigator at Cedars-Sinai and first author of the paper.

Arditi,theGUESS?/Fashion Industries Guild Chair in Community Child Healthat Cedars-Sinai who leads theInfectious and Immunologic Diseases Research Center,and colleagues are currently conducting a study in which they are analyzing cerebral spinal fluid samples from people with long COVID symptoms for evidence of neurotoxic motifs.

Rebecca A. Porritt, PhD, assistant professor in the departments of Pediatrics and Biomedical Sciences at Cedars-Sinai, also contributed to this work.

Funding: The study was funded by the National Institutes of Health (award numbersR01AI072726, R01AI072726-10S, GM103712, R01GM139297, R01HL139766 and R01HL159297) the American Heart Association (Career Development Award AHA 20CDA35260258), and the Cedars-Sinai Precision Health Award.

Frontiers in Immunology

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.

See the original post here:
Hyperactivation of the immune system may cause post-COVID syndromes - EurekAlert

Read More...

Page 9«..891011..2030..»


2024 © StemCell Therapy is proudly powered by WordPress
Entries (RSS) Comments (RSS) | Violinesth by Patrick