A study of around 500,000 medical records has suggested that severe viral infections like encephalitis and pneumonia increase the risk of neurodegenerative diseases like Parkinson’s and Alzheimer’s.
Amyloid plaques in Alzheimer’s disease (Kateryna Kon/Science Photo Library/Getty Images)
A study of around 500,000 medical records has suggested that severe viral infections like encephalitis and pneumonia increase the risk of neurodegenerative diseases like Parkinson’s and Alzheimer’s.
Researchers found 22 connections between viral infections and neurodegenerative conditions in the study of around 450,000 people.
People treated for a type of inflammation of the brain called viral encephalitis were 31 times more likely to develop Alzheimer’s disease. (For every 406 viral encephalitis cases, 24 went on to develop Alzheimer’s disease – around 6 percent.)
Those who were hospitalized with pneumonia after catching the flu seemed to be more susceptible to Alzheimer’s disease, dementia, Parkinson’s disease, and amyotrophic lateral sclerosis (ALS).
Intestinal infections and meningitis (both often caused by a virus), as well as the varicella-zoster virus, which causes shingles, were also implicated in the development of several neurodegenerative diseases.
The impact of viral infections on the brain persisted for up to 15 years in some cases. And there were no instances where exposure to viruses was protective.
Around 80 percent of the viruses implicated in brain diseases were considered ‘neurotrophic’, which means they could cross the blood-brain barrier.
“Strikingly, vaccines are currently available for some of these viruses, including influenza, shingles (varicella-zoster), and pneumonia,” the researchers write.
“Although vaccines do not prevent all cases of illness, they are known to dramatically reduce hospitalization rates. This evidence suggests that vaccination may mitigate some risk of developing neurodegenerative disease.”
“After reading [this] study, we realized that for years scientists had been searching – one-by-one – for links between an individual neurodegenerative disorder and a specific virus,” said senior author Michael Nalls, a neurogeneticist at the National Institute on Aging in the US.
“That’s when we decided to try a different, more data science-based approach,” he said. “By using medical records, we were able to systematically search for all possible links in one shot.”
First, the researchers analyzed the medical records of around 35,000 Finns with six different types of neurodegenerative diseases and compared this against a group of 310,000 controls who did not have a brain disease.
This analysis yielded 45 links between viral exposure and neurodegenerative diseases, and this was narrowed down to 22 links in a subsequent analysis of 100,000 medical records from the UK Biobank.
While this retrospective observational study cannot demonstrate a causal link, it adds to the pile of research hinting at the role of viruses in Parkinson’s and Alzheimer’s disease.
“Neurodegenerative disorders are a collection of diseases for which there are very few effective treatments and many risk factors,” said co-author Andrew Singleton, a neurogeneticist and Alzheimer’s researcher and the director of the Center for Alzheimer’s and Related Dementias.
“Our results support the idea that viral infections and related inflammation in the nervous system may be common – and possibly avoidable – risk factors for these types of disorders.”
Alzheimer’s disease is a devastating degenerative brain condition that affects millions of people in the U.S. While pharmaceutical treatments have long lists of side effects, there is a natural food-based intervention that has proven effective in improving key brain functions. The best part is, it’s probably in your food pantry right now
If you’re a regular reader of GreenMedInfo.com, you’re likely to have seen numerous articles detailing the dozens of healthy uses for coconut oil that are backed by science. From balancing blood sugar[i] and hormones[ii] to healing burns[iii] and ulcers,[iv] it seems there is hardly an ailment that is not soothed or supported by adding this nutrition-dense fat to your diet.
Coconut Oil: The Brain’s Preferred Fuel?
In 2018, researchers added to the knowledge base with confirmation of coconut oil’s usefulness as a brain-boosting superfood. The pilot study,[v]published in July 2018 in the Journal of Alzheimer’s Disease, has shown that a Mediterranean diet, rich in coconut oil, improves the main cognitive functions in patients with Alzheimer’s disease (AD).
Conducted by a multidisciplinary team of researchers from the Catholic University of Valencia, Spain, the aim of the study was to detect changes in key cognitive functions of patients with AD after following a traditional Mediterranean diet boosted by therapeutic doses of coconut oil.
Study methods were prospective, longitudinal, qualitative and analytic, meaning participants’ health and behaviors were studied across time to observe unknown and unpredicted changes in outcomes. Inclusion criteria were diagnosed AD patients, aged 65 to 85 years old, who were institutionalized in the Alzheimer’s Family Association of Valencia (AFAV).
A representative sample size of 44 participants was ultimately selected from the original pool of 458 AFAV patients, with criteria excluding patients who were diagnosed with other types of degenerative cognitive disorder or verbal disability that prevented them from answering test questions, and excluding patients with any metabolic chronic disease or who had been treated with drugs such as antidepressants, antipsychotics or hypnotic drugs, which could alter cognitive functions.
The 44 participants were randomly divided into two homogenous groups comprised of 22 patients each: an experimental group receiving coconut oil supplementation and a control group that did not receive coconut oil. Both groups followed an isocaloric Mediterranean diet that was shown in previous studies to be associated with a decrease in cognitive impairment in AD patients.
In the Mediterranean diet implemented in this study, proteins accounted for 15% of total calories, carbohydrates for 55% and lipids for 30% of overall energy intake. Calorie intake was the same for all participants, taking into account that in the experimental group, lipids were reduced so that by adding the coconut oil supplement, the daily lipid amount for all study participants was the same. The dietary intervention was conducted over a period of 21 days.
Cognitive changes in participants were measured by the same institutional psychologist, blind to study protocols, who conducted the “7-Minute Screen,” an assessment that measures “temporal orientation, visuospatial and visuoconstructive abilities.” Visuoconstructive disabilities are represented by difficulty doing math, driving and writing, among other common daily tasks. Patients were assessed the day before dietary therapy and the day after therapy throughout the 21-day intervention.
Alzheimer’s: Most Prevalent Brain Disorder
According to researchers, “Alzheimer’s disease (AD) is the most prevalent neurodegenerative disorder, and new therapies are needed.” This study was a method of proving their hypothesis that coconut oil can be a beneficial source of ketone bodies, an alternative brain fuel to glucose for AD patients whose hypometabolism, or poor glucose utilization, is a factor in their disease.
In addition to serving as a fuel source for brain metabolism, ketone bodies, in adequate doses, regulate glutamate release in the synaptic cleft, the space between neurons that transmits impulses.[vi]Glutamate is a neurotransmitter that is involved in most aspects of normal brain function.[vii]
Researchers stated that gender is a significant factor in AD, with women more commonly affected than men.
The trial groups had 75% female and 25% male patients, reflecting the same percentages of gender distribution as the disease itself.[viii] In the published paper, the scientists noted the insidious onset of Alzheimer’s disease, which initially becomes apparent through “progressive loss of episodic memory, followed by gradual impairment of declarative and non-declarative memory.
Later, loss of other main cognitive functions, such as language, executive functions, attention span, and working memory, have also been observed as well as alterations in temporal orientation, visuospatial ability, and visuoconstructive ability.”[ix]
After a baseline assessment of all participants using the 7-Minute Screen, dosing commenced consisting of 20 milliliters (mls) of coconut oil, twice daily, for a total daily dose of 40 mls. This amount of coconut oil had previously demonstrated effectiveness at improving cognitive functions over 21 days in human[x] and mice studies.[xi]
Coconut Oil Improves Information Processing and Memory in AD Patients
Results were both confirming of the researchers’ hypothesis regarding the benefits of coconut oil and encouraging for proponents of natural disease interventions:
“Taking a closer look at the changes observed in the group that received coconut oil, these changes seem to point to the fact that certain cognitive functions improved … such as temporal orientation (information processing), semantic memory and episodic memory …
[These improvements] … could be explained by the decrease in insulin resistance due to the action of ketone bodies, since memory improvement has been observed after intranasal administration of insulin in AD patients, which increases glucose metabolism.”[xii]
An important observation was made regarding the potential for brain recovery with coconut oil: “It could be deduced that not all regions of the cerebral cortex recover to the same degree.”[xiii]Regarding gender differences, researchers observed that “female patients recover more easily than male patients, which confirms our previous results, where a global cognitive improvement was shown in women.”[xiv]
They hypothesize that these results could possibly be explained by hormonal differences in sex, “but not only with respect to low estrogen levels but also … by testosterone, whose levels of production are much lower in women with AD and cause them to have higher insulin resistance.”[xv] Researchers concluded that the positive effects of coconut oil are not gender- or state-specific, however, the benefits are “more evident in women with mild-moderate state [AD].”
Final conclusions of the study were that an isocaloric, coconut oil-enriched Mediterranean diet improves cognitive functions in patients with AD, with differences according to patient sex and degree of severity of the disease.[xvi] They issued a call for further studies of this type to add to this important body of evidence.
To learn more about the health benefits of coconut oil, GreenMedInfo.com has more than 70 abstracts in the world’s most widely referenced natural health database.
References
[i] Protective and Antidiabetic Effects of Virgin Coconut Oil (Vco) on Blood Glucose Concentrations in Alloxan Induced Diabetic Rats. Nur‘azimatul Quddsyiah H. Maidin, Norhayati Ahmad. International Journal of Pharmacy and Pharmaceutical Sciences. Vol 7, Issue 10, 2015. ISSN: 0975-1491. https://pdfs.semanticscholar.org/62d6/b586d89f623b4be84ac93c828b31f1070b76.pdf
[ii] Effect of dietary saturated fatty acids on hormone-sensitive lipolysis in rat adipocytes. Awad AB, Chattopadhyay JP. J Nutr. 1986 Jun;116(6):1088-94. PMID: 3014093
[iii] Burn wound healing property of Cocos nucifera: An appraisal. Srivastava P, Durgaprasad S. Indian J Pharmacol. 2008 Aug;40(4):144-6. doi: 10.4103/0253-7613.43159. PMID: 20040946
[v]Improvement of Main Cognitive Functions in Patients with Alzheimer’s Disease after Treatment with Coconut Oil Enriched Mediterranean Diet: A Pilot Study. de la Rubia Ortí JE, et al. J Alzheimers Dis. 2018;65(2):577-587. doi: 10.3233/JAD-180184. PMID: 30056419
[vii] Improvement of Main Cognitive Functions in Patients with Alzheimer’s Disease after Treatment with Coconut Oil Enriched Mediterranean Diet: A Pilot Study. de la Rubia Ortí JE, et al. J Alzheimers Dis. 2018;65(2):577-587. doi: 10.3233/JAD-180184. PMID: 30056419
[viii] Improvement of Main Cognitive Functions in Patients with Alzheimer’s Disease after Treatment with Coconut Oil Enriched Mediterranean Diet: A Pilot Study. de la Rubia Ortí JE, et al. J Alzheimers Dis. 2018;65(2):577-587. doi: 10.3233/JAD-180184. PMID: 30056419
[ix] Lazarov O, Hollands C. Hippocampal neurogenesis: Learning to remember. Prog Neurobiol. 2016;138-140:1–18. doi:10.1016/j.pneurobio.2015.12.006. PMID: 26855369
[x]Farah BA (2014) Effects of caprylic triglyceride on cognitive performance and cerebral glucose metabolism in mild Alzheimer’s disease: A single-case observation. Front Aging Neurosci 16, 1-4. PMID: 25076901
[xi] Reger MA, Henderson ST, Hale C, Cholerton B, Baker LD, Watson GS, Hyde K, Chapman D, Craft S (2004) Effects of beta-hydroxybutyrate on cognition in memory-impaired adults. Neurobiol Aging 25, 311-314. PMID: 15123336
[xii] How does coconut oil affect cognitive performance in alzheimer patients? de la Rubia Ortí JE, et al. Nutr Hosp. 2017 Mar 30;34(2):352-356. doi: 10.20960/nh.780. PMID: 28421789
[xiii] Improvement of Main Cognitive Functions in Patients with Alzheimer’s Disease after Treatment with Coconut Oil Enriched Mediterranean Diet: A Pilot Study. de la Rubia Ortí JE, et al. JAlzheimers Dis. 2018;65(2):577-587. doi: 10.3233/JAD-180184. PMID: 30056419
[xiv] Improvement of Main Cognitive Functions in Patients with Alzheimer’s Disease after Treatment with Coconut Oil Enriched Mediterranean Diet: A Pilot Study. de la Rubia Ortí JE, et al. J Alzheimers Dis. 2018;65(2):577-587. doi: 10.3233/JAD-180184. PMID: 30056419
[xv] Improvement of Main Cognitive Functions in Patients with Alzheimer’s Disease after Treatment with Coconut Oil Enriched Mediterranean Diet: A Pilot Study. de la Rubia Ortí JE, et al. J Alzheimers Dis. 2018;65(2):577-587. doi: 10.3233/JAD-180184. PMID: 30056419
[xvi] Improvement of Main Cognitive Functions in Patients with Alzheimer’s Disease after Treatment with Coconut Oil Enriched Mediterranean Diet: A Pilot Study. de la Rubia Ortí JE, et al. J Alzheimers Dis. 2018;65(2):577-587. doi: 10.3233/JAD-180184. PMID: 30056419
The GMI Research Group (GMIRG) is dedicated to investigating the most important health and environmental issues of the day. Special emphasis will be placed on environmental health. Our focused and deep research will explore the many ways in which the present condition of the human body directly reflects the true state of the ambient environment.
Disclaimer: This article is not intended to provide medical advice, diagnosis or treatment. Views expressed here do not necessarily reflect those of GreenMedInfo or its staff.
(CNN)Authorities in China have approved a drug for the treatment of Alzheimer’s disease, the first new medicine with the potential to treat the cognitive disorder in 17 years.
The seaweed-based drug, called Oligomannate, can be used for the treatment of mild to moderate Alzheimer’s, according to a statement from China’s drug safety agency. The approval is conditional however, meaning that while it can go on sale during additional clinical trials, it will be strictly monitored and could be withdrawn should any safety issues arise.
In September, the team behind the new drug, led by Geng Meiyu at the Shanghai Institute of Materia Medica under the Chinese Academy of Sciences, said they were inspired to look into seaweed due to the relatively low incidence of Alzheimer’s among people who consume it regularly.
In a paper in the journal Cell Research, Geng’s team described how a sugar contained within seaweed suppresses certain bacteria contained in the gut which can cause neural degeneration and inflammation of the brain, leading to Alzheimer’s.
This mechanism was confirmed during a clinical trial carried out by Green Valley, a Shanghai-based pharmaceutical company that will be bringing the new drug to market.
Conducted on 818 patients, the trial found that Oligomannate — which is derived from brown algae — can statistically improve cognitive function among people with Alzheimer’s in as little as four weeks, according to a statement from Green Valley.
“These results advance our understanding of the mechanisms that play a role in Alzheimer’s disease and imply that the gut microbiome is a valid target for the development of therapies,” neurologist Philip Scheltens, who advises Green Valley and heads the Alzheimer Center Amsterdam, said in the statement.
Vincent Mok, who heads the neurology division at the Chinese University of Hong Kong, said the new drug showed “encouraging results” when compared to acetylcholinesterase inhibitors — the existing treatment for mild to severe Alzheimer’s.
“It is just as effective but it has fewer side effects,” he told CNN. “It will also open up new avenues for Alzheimer’s research, focusing on the gut microbiome.”
Alzheimer’s disease is one of the most feared illnesses afflicting older adults. Just ask anyone over the age of 55 who has misplaced car keys whether they didn’t have a momentary fear it could be a first sign of the crippling slow decline associated with this disease.
Most of what Doctors diagnose as “dementia” is Alzheimer’s disease (about 70% of dementia cases). With our rapidly aging populations, Alzheimer’s has become the 5th leading cause of death in the U.S. As a result, many of us have witnessed the heartbreak and despair of watching a loved one’s progressive memory loss and brain function resulting in the eventual but often slow death of the person they once knew. We watch our family members disintegrate in slow motion before our eyes and pray that we never meet the same fate.
Throughout the years, a lot of Alzheimer’s research focused on how the characteristic amyloid deposits on the brain are formed and exploring the role of inflammation in the disease progression. These amyloid proteins form large sticky plaques on the brain and are a hallmark of the disease and an early indicator of Alzheimer’s.
But finally, after decades of scientific failure and billions of dollars spent in looking for a cure along with a 99% failure rate in drug treatment research, there is a bright ray of hope that may lead to a long-sought-after treatment.
For several years now, research has identified a relationship between gum disease in older patients and the presence of amyloid beta plaques characteristic of Alzheimer’s disease in the brains of its sufferers.1 Before this line of investigation, the chronic periodontal disease found in Alzheimer’s patients was often dismissed as merely the result of dementia for those who could no longer maintain good oral hygiene.
But, this periodontal disease connection intrigued an international team of scientists and they decided to take a more in-depth look at the role of gum disease in Alzheimer’s. And they have published some truly intriguing results.
It was a complex series of studies worked on by several groups of scientists. So I’m going to try and break it down for you: First, they analyzed cerebrospinal fluid (considered a “window” into brain infection) from 10 living patients diagnosed with probable Alzheimer’s disease. Then they isolated DNA from Porphyromonas gingivalis (Pg), the primary pathogen found in chronic periodontitis, matching it with the Pg that the researchers found in the patients’ saliva samples.2
Next, the researchers looked at the toxic enzymes produced by Pg called “gingipains,” and analyzed brain-bank samples from deceased people. One team found that there were greater gingipain loads in the brains of people who died from Alzheimer’s disease than in the brains of people who had no diagnosis of dementia.3
This new information quickly led them to the conclusion that Pg “is not a result of poor dental care following the onset of dementia or a consequence of late-stage disease, but is an early event that can explain the pathology found in middle-aged individuals before cognitive decline.”2
Now for the fascinating part of the study. The team tested a molecular therapy already undergoing clinical trials on Alzheimer’s patients to see if the compound, called COR388, could inhibit the toxic action of gingipains in the brains of mice that had been orally infected with gingipains. They found that not only was there reduced bacterial load of Pg brain infection, COR388 also blocked amyloid beta production, and reduced neuroinflammation.
Overall this added up to more protection of neurons in the hippocampus—the part of the brain that controls memory and is damaged early in the development of Alzheimer’s.4
What Does This Mean For Everyday People?
Dr. Jan Potempa, an investigator based at the University of Louisville’s Department of Oral Immunology and Infections Disease who was part of the international team said, “We now have strong evidence connecting P. gingivalis and Alzheimer’s pathogenesis (the chain of events leading to the disease)…”
An even more critical aspect of this study is that there might be enormous potential for therapies that could change the course of the disease or even stop the progression of a disease that now seems to be strongly connected with this particular dental disease and bacteria.4 Researchers are now focusing on developing a compound like COR388 that can effectively block the debilitating effects of Pg in the brain.
Also, consider this, there is a link between gum disease and other conditions such as heart disease and diabetes. The research here is also not conclusive, but there is some evidence that suggests that there is a connection and more research is underway.
According to The Mayo Clinic5 :
“Gum disease (periodontitis) is associated with an increased risk of developing heart disease.”
“Poor dental health increases the risk of a bacterial infection in the bloodstream, which can affect the heart valves. Oral health may be particularly important if you have artificial heart valves.”
“Tooth loss patterns are connected to coronary artery disease.
“There is a strong connection between diabetes and cardiovascular disease and evidence that people with diabetes benefit from periodontal treatment.”
Bottom line: Though this line of investigation is still young and COR388 may not prove to be a magic bullet (or the only magic bullet) against Alzheimer’s disease, this research is very encouraging, and we will continue to update this article with any new studies. Until then, it certainly makes sense to maintain good oral hygiene practices (regular brushing and flossing) and visit a dentist regularly to catch gum disease as early as possible!
2Dominy SS et al. Porphyromonas gingivalis in Alzheimer’s disease brains: Evidence for disease causation and treatment with small-molecule inhibitors.Science Advances. 2019. http://advances.sciencemag.org/content/5/1/eaau3333
Antidepressants and antihistamines are among the most common types of medications people take, and they belong to a class of drugs known as anticholinergics. These drugs can treat a variety of health problems, including COPD, asthma, depression, dizziness, gastrointestinal problems, overactive bladder, and the symptoms of Parkinson’s. Although they can be effective, a large new study has shown that if you take them, you might just be trading one problem for another, possibly bigger one: dementia.
Although people who suffer from depression may be desperate to get relief from this illness that can have such a negative impact on daily life, tricyclic antidepressants fall into this category, so it’s important to pay attention the concerning new findings if you take medications like Elavil, Deptran, Sinequan, or Silenor. The same can be said for antihistamines like Benadryl, among other drugs.
The study, which was published in BMJ, involved more than 40,000 dementia patients and more than 283,000 people who don’t have dementia and followed them from 2006 to 2015. They found that people who had dementia had a greater likelihood of having taken class 3 anticholinergic drugs prior to developing the illness.
These medications block the actions of acetylcholine in the brain, which can prevent it from causing involuntary movements in the muscles in the lungs, urinary tract, gastrointestinal tract, and other parts of the body.
Although the higher risk varied depending on the drugs, some of them raised the risk by 30 percent. Not every anticholinergic drug had the effect, but using some of them even as far back as 20 years raised a person’s risk of dementia later on. Generally speaking, they believe that a person aged 65 to 70 sees their risk of dementia increase by 19 percent if they’ve used anticholinergic antidepressants. The association with dementia goes up with greater levels of exposure to the meds.
The study was praised by experts for its strength and using U.K. healthcare databases rather than relying on patient recall, which isn’t always dependable.
The drugs are believed to have this effect because anticholinergic medications lower the levels of a chemical called acetylcholine in the brain, which is a crucial messenger in memory pathways. This is a known effect that already stops some doctors from prescribing such drugs to older and more frail patients.
Other studies have reached a similar conclusion about anticholinergic drugs
In a different study involving nearly 3,500 people, researchers reached a similar conclusion, finding that those who used anticholinergic drugs had a greater likelihood of developing dementia, and their risk increased according to their cumulative dose. For example, taking such meds for three years or longer was linked to a 54 percent rise in dementia risk compared to taking the same dose for less than three months.
Experts say such findings are a good reminder that people should evaluate all the medications they’re taking from time to time to see if they are really working for you. For example, if you’re taking antidepressants and are still depressed, the medications may not be helping. Many of these drugs have safer alternatives, including non-medication approaches that could make a difference safely and effectively.
With the number of people suffering from Alzheimer’s expected to triple by 2050, it’s important to do all you can to minimize your risk – and that includes staying away from anticholinergic drugs if possible.
Alzheimer’s mental decline can be headed off years in advance of the occurrence of symptoms with the use of vitamin C supplements but not with oranges or apples that are considered to be vitamin C-rich foods.
Don’t wait for your doctor or public health authorities to produce a vaccine or a drug to prevent the predicted tripling of Alzheimer’s disease from 4.7 to 13.8 million by the year 2050. The research community has already identified the initial step in the development of Alzheimer’s but is reluctant to make a public health pronouncement that would upset the current pharmaceutical drug paradigm that predominates in modern medicines (none of the five classes of approved drug for this brain disease address its cause).
Mounting evidence points to vitamin C as a preventive measure to head off this ongoing mental health catastrophe. Consumption of vitamin C-rich foods (example: oranges) won’t be effective. Dietary supplements would be needed and taken throughout the day to achieve optimal blood levels as this water-soluble nutrient is rapidly excreted.
It has been known since the 1940s that vitamin C strengthens blood capillaries. Capillaries are connectors between the red hoses (arteries) that carry oxygenated blood and nutrients to tissues and the blue hoses (vein) used to dispose of deoxygenated blood.
Prevention, can’t afford treatment
Worldwide Alzheimer’s cases are predicted to jump from 50 to 152 million over the same time. Annual cost of care is $818 billion/year now and will rise to $2 trillion, stifling world economies and in whispers, forcing health organizations to horrifically think of ways of covertly culling the demented population. That is what public health authorities are saying will happen “unless preventive measures are developed.”
(Natural News) Americans walk, run, and march “for the cure” for all kinds of different diseases, helping to raise awareness and funds for research, but what if you found out right now there’s a cure for Alzheimer’s Disease, would you “take care of business” starting now or keep wishing someone else might come along to possibly save you later?
Sure, right about now you’re hoping the cure will come in some magical pill or prolific injection, and do the job “overnight,” so you won’t have to do any work or garner long-term diligence – well, there’s good news and bad news – and they’re both the same. Scientists have figured out what causes Alzheimer’s Disease and what cures it, but it’s not some chemical pill or experimental vaccine, so let’s get to work.
Understanding the neuroscience of Alzheimer’s and Parkinson’s shows us the cause and the cure at the same time
The point of connection of neurons is called a synapse, and that’s where neurotransmitters are released and communication happens in the brain. This is where we experiences all of our senses and engage in thought processes, including critical thinking and memory. This is also exactly where dementia happens.
The synapse is where neurons release hormones, glutamates, and small peptides called amyloid beta. The amyloid beta are the brain’s “trash” and a prime factor involved in Alzheimer’s disease, functioning as the main component of plaques that cling to each other and clog up the neural pathway. These are the plaques found in the brains of Alzheimer’s patients.
Normally, these amyloid plaques are swept out of the neural pathway (like trash) by the “custodians of the brain” called microglea. These amazing microglea are the brain’s own immune cells and are the answer to beating brain diseases. Scientists recently discovered through sophisticated experiments that these cells constantly search for brain damage, like a perpetually-running computer virus scan, running surveillance for different levels of damage. The microglea are literally capable of eating infected and damaged cells before infection spreads, while clearing out “debris” from dying cells.
Diseases of dementia therefore begin when amyloid beta begins to accumulate, because too much is released, overwhelming the microglea, and leaving waste in the neural pathways, blocking communication. The synapse piles up with plaques (trash and waste) that become sticky and bind to themselves (think of animal fat clogging your sink drain).
At a certain tipping point, when the body and brain have created too much “trash” for too long, creating massive inflammation and tangles, the microglea become overwhelmed and enter a hyper-mode, where they actually begin attacking healthy cells. Scientists believe the microglea may even, at the tipping point, begin clearing away the synapses themselves. Get it? The cure lives in keeping amyloid plaques from reaching the “tipping point.” Here’s how you do that.
Stop consuming foods that create plaques in the brain – so your brain’s “custodians” can clear out the sticky trash that blocks your synapses
Amyloid plaque accumulation may never be “cured” with a chemical drug or vaccine, but that doesn’t matter, because you can cure the problem yourself. Are you ready to start taking your preventative medicine? It’s not very difficult you know. Let’s break it down to its simplest form, then you decide if you can “pull it off.”
You wouldn’t pick up a poisonous snake just to see if it bites you, and then start searching the internet for the anecdote, would you? You wouldn’t pick some poison ivy and rub it on your skin on purpose, would you? If you were severely allergic to peanuts, you certainly wouldn’t eat a handful just to see what happens. That’s just common sense.
So what if you knew what caused dementia, would you stop eating it? Guess what. Now is the time to stop marching for the cure and start living it, because knowledge is power. Now get this.
White foods are known to cause excess plaque build-up in the brain, leading to dementia. These white foods include white bread, white flour, white rice (except basmati, which is naturally white), white pasta, and white sugar. Stop eating bleached food.
Processed foods and meats cause excess plaque in the synapses, fueling dementia. Avoid processed cheeses (think American cheese especially here), and processed meats, like sausages, bacon, hot dogs, and cold cuts (especially smoked deli meats), and even beer. Nitrosamines in smoked meats cause the liver to produce fats that are toxic to the brain.
Stop eating foods that contain diacetyl, a chemical commonly found in microwave popcorn. Diacetyl increases amyloid plaques in the brain.
Animal fat and canola oil coagulate in your blood and create tangles of plaque in the brain
You’ll hear it time and time again, that a plant-based diet cures almost every preventable disease and disorder known to humans. It’s true. If you’re a heavy meat eater, your body is struggling to process all that animal fat, creating heart and brain “trash” that your body’s “janitors” just can’t sweep away fast enough.
If you think organic or “expeller pressed” canola oil means that the oil doesn’t coagulate in your body, you’d be dead wrong. After about six weeks, any canola oil that your body hasn’t cleared out looks like a sticky glue you could use to bond cement. Think of all that “trash” blocking your synapses and causing dementia, because that’s exactly what happens.
Did you know that in the U.S. alone, Alzheimer’s care already costs $2 billion a year (one out of every five Medicare dollars)? Dementia kills more people than cancer. Did you know that? Sure, Big Pharma will tell you Alzheimer’s and Parkinson’s are not preventable, but both are, and the cure lives in prevention. You may begin now.
A new study links poor sleep quality in older adults with elevated levels of tau, a protein associated with Alzheimer’s disease. Researchers report poor sleep quality later in life may be associated with declining brain health and may be an early indicator of Alzheimer’s disease.
Summary: A new study links poor sleep quality in older adults with elevated levels of tau, a protein associated with Alzheimer’s disease. Researchers report poor sleep quality later in life may be associated with declining brain health and may be an early indicator of Alzheimer’s disease.
Source: WUSTL.
Poor sleep is a hallmark of Alzheimer’s disease. People with the disease tend to wake up tired, and their nights become even less refreshing as memory loss and other symptoms worsen. But how and why restless nights are linked to Alzheimer’s disease is not fully understood.
Now, researchers at Washington University School of Medicine in St. Louis may have uncovered part of the explanation. They found that older people who have less slow-wave sleep – the deep sleep you need to consolidate memories and wake up feeling refreshed – have higher levels of the brain protein tau. Elevated tau is a sign of Alzheimer’s disease and has been linked to brain damage and cognitive decline.
The findings, published Jan. 9 in Science Translational Medicine, suggest that poor-quality sleep in later life could be a red flag for deteriorating brain health.
“What’s interesting is that we saw this inverse relationship between decreased slow-wave sleep and more tau protein in people who were either cognitively normal or very mildly impaired, meaning that reduced slow-wave activity may be a marker for the transition between normal and impaired,” said first author Brendan Lucey, MD, an assistant professor of neurology and director of the Washington University Sleep Medicine Center. “Measuring how people sleep may be a noninvasive way to screen for Alzheimer’s disease before or just as people begin to develop problems with memory and thinking.”
The brain changes that lead to Alzheimer’s, a disease that affects an estimated 5.7 million Americans, start slowly and silently. Up to two decades before the characteristic symptoms of memory loss and confusion appear, amyloid beta protein begins to collect into plaques in the brain. Tangles of tau appear later, followed by atrophy of key brain areas. Only then do people start showing unmistakable signs of cognitive decline.
The challenge is finding people on track to develop Alzheimer’s before such brain changes undermine their ability to think clearly. For that, sleep may be a handy marker.
To better understand the link between sleep and Alzheimer’s disease, Lucey, along with David Holtzman, MD, the Andrew B. and Gretchen P. Jones Professor and head of the Department of Neurology, and colleagues studied 119 people 60 years of age or older who were recruited through the Charles F. and Joanne Knight Alzheimer’s Disease Research Center. Most – 80 percent – were cognitively normal, and the remainder were very mildly impaired.
The researchers monitored the participants’ sleep at home over the course of a normal week. Participants were given a portable EEG monitor that strapped to their foreheads to measure their brain waves as they slept, as well as a wristwatch-like sensor that tracks body movement. They also kept sleep logs, where they made note of both nighttime sleep sessions and daytime napping. Each participant produced at least two nights of data; some had as many as six.
The researchers also measured levels of amyloid beta and tau in the brain and in the cerebrospinal fluid that bathes the brain and spinal cord. Thirty-eight people underwent PET brain scans for the two proteins, and 104 people underwent spinal taps to provide cerebrospinal fluid for analysis. Twenty-seven did both.
After controlling for factors such as sex, age and movements while sleeping, the researchers found that decreased slow-wave sleep coincided with higher levels of tau in the brain and a higher tau-to-amyloid ratio in the cerebrospinal fluid.
“The key is that it wasn’t the total amount of sleep that was linked to tau, it was the slow-wave sleep, which reflects quality of sleep,” Lucey said. “The people with increased tau pathology were actually sleeping more at night and napping more in the day, but they weren’t getting as good quality sleep.”
Reduced amounts of slow brain waves – the kind that occur in deep, refreshing sleep – are associated with high levels of the toxic brain protein tau. This computer-generated image maps the areas where the link is strongest, in shades of red and orange. A new study from Washington University School of Medicine in St. Louis has found that decreased deep sleep is associated with early signs of Alzheimer’s disease. NeuroscienceNews.com image is credited to Brendan Lucey.
If future research bears out their findings, sleep monitoring may be an easy and affordable way to screen earlier for Alzheimer’s disease, the researchers said. Daytime napping alone was significantly associated with high levels of tau, meaning that asking a simple question – How much do you nap during the day? – might help doctors identify people who could benefit from further testing.
“I don’t expect sleep monitoring to replace brain scans or cerebrospinal fluid analysis for identifying early signs of Alzheimer’s disease, but it could supplement them,” Lucey said. “It’s something that could be easily followed over time, and if someone’s sleep habits start changing, that could be a sign for doctors to take a closer look at what might be going on in their brains.”
Contrary to conventional wisdom, brain regeneration is possible. One promising therapy that promotes neurogenesis and is effective in pre-clinical studies of Alzheimer’s and Parkinson’s is near infrared light therapy, and it may improve other mental illnesses and neurodegenerative disorders including dementia, stroke, ALS, and traumatic brain injury as well.
Alzheimer’s disease and Parkinson’s disease are the most common neurodegenerative disorders. The former is a type of dementia that occurs secondary to the accumulation of abnormal protein deposits in the brain, including β-amyloid plaques and intraneuronal neurofibrillary tangles made of tau protein (1). Upon neuroimaging studies, gross cerebral cortical atrophy is found, meaning that the part of the brain responsible for executive functions such as learning, memory, language, decision-making, and problem-solving progressively degenerates (1). In addition, gliosis, or brain inflammation, is a hallmark characteristic of Alzheimer’s (1).
One hypothesis that is championed proposes that Alzheimer’s occurs due to self-propagating, prion-like protein assemblies, which interfere with the function of nerve cells (2). An alternate theory is that these so-called proteinopathies occur secondary to a microvascular hemorrhage or brain bleed (3). The brain bleed is believed to be the result of age-induced degradation of cerebral capillaries, which creates neuron-killing protein plaques and tangles (3).
Dysfunction of mitochondria, the energy-generating powerhouses of the cell, is also implicated in Alzheimer’s, as reduced efficacy of these organelles creates oxidative stress-inducing reactive oxygen species, or free radicals, which lead to neuronal cell death (4). Whatever the cause, extensive death of brain cells occurs, which explains the cognitive deficits that occur with Alzheimer’s disease, in addition to symptoms such as impaired judgment, confusion, agitation, linguistic abnormalities, social withdrawal, and even hallucinations (1).
Parkinson’s disease, on the other hand, is characterized by progressive death of dopamine-producing neurons in a region of the brainstem called the substantial nigra, but it can extend to other brain areas such as the locus coeruleus, olfactory bulb, dorsal motor nucleus of the vagal nerve, and even the cortex in late stages (5). As a result, the primary manifestation is that dopamine deficiency appears in the basal ganglia, a set of nuclei embedded deep in the brain hemispheres that is responsible for motor control (6). This leads to the cardinal manifestation of Parkinson’s, namely, a movement disorder that includes bradykinesia or slow movement, loss of voluntary movement, muscular rigidity, and resting tremor (7).
Not unlike what happens in Alzheimer’s, accumulation of abnormal intracellular protein aggregates known as Lewy bodies, composed of a protein called α-synuclein, is thought to be central to the pathogenesis of Parkinson’s disease (8). Like Alzheimer’s, mitochondrial dysfunction induced by genetic mutations, toxic agents, or damage to blood vessels is also considered to contribute to neuron cell death in Parkinson’s (9). Toxin exposure is especially implicated, as animal studies hint that development of Parkinson’s disease may occur as a byproduct of exposure to neurotoxins such as rotenone or paraquat (10). Impaired blood brain barrier function and damage to the endothelial cells of the vascular system, which line the interior surface of blood vessels, are also thought to play a role in Parkinson’s (10).
Overturning Old Notions of Neuroscience
The central dogma of neuroscience conceived of the central nervous system tissue as “perennial” after the doctrines of Giulio Bizzozero, the most prominent Italian histologist, who decreed that the lifelong cells of the nervous system were devoid of replicative potential (11). In other words, the perennial nature ascribed to the nerve cells of the brain and spinal cord meant that nerve cells were believed to be incapable of undergoing proliferation, or cell division, in the postnatal brain (11). While the early stage of in utero prenatal development known as embryogenesis permits massive neurogenesis, or the ability to create new nerve cells, the scientific consensus up until the end of the twentieth century held that neurogenesis was arrested after birth in mammals.
Santiago Ramon y Cajal, who led the charge in the neuroscience discipline in the later half of the nineteenth century onward and won a Nobel Prize for Medicine and Physiology, in fact stated that: “Once development was ended, the fonts of growth and regeneration of the axons and dendrites dried up irrevocably. In adult centers, the nerve paths are something fixed and immutable: everything may die, nothing may be regenerated” (11). Acknowledgment of the mere possibility of adult neurogenesis was hampered by the fact that scientists lacked the visualization techniques to detect neural stem cells, the precursors to new neurons and means by which neurogenesis occurs, and also did not have access to the molecular markers and microscopy required to observe cells in different cycle phases.
This view of nervous tissue as perennial was also reinforced by clinical observations that patients with chronic neurodegeneration, traumatic brain lesions, and cerebrovascular diseases do not experience functional recovery (11). Prevailing theories posited that adult neurogenesis was an evolutionary unlikelihood, since it would interfere with pre-existing neuronal connections and the fine-tuned electrochemical communication in the nervous system, as well as disrupt memory recall, which was believed to occur via stable neuronal circuits created and encoded during learning (11).
That brain cells are finite, and incapable of regeneration, painted a portrait of doom and gloom and inexorable debilitation for patients suffering from devastating neurodegenerative conditions. However, relatively recent discoveries have overturned these antiquated conceptions by revealing that the brain is plastic, or pliable, and that even neurons in adult higher vertebrates are capable of neurogenesis.
Scientists Discover Neural Regeneration is Possible
In the 1960s, these postulates of the old neurobiology were disproven when Joseph Altman and colleagues performed an experiment where radioactively labelled thymidine, one of the nucleotide base pairs that makes up DNA, was incorporated into a brain area called the dentate gyrus of the hippocampus and integrated into the genetic material of what was later confirmed via electron microscopy to be dividing neurons (12, 13). In essence, this illustrated that neurons were undergoing mitosis, a process of cell division where genetically identical daughter cells are created, and showed that adult neurogenesis is possible.
Another nail in the coffin of this antiquated perception of the nervous system was that neural stem cells, the multipotent, self-renewing progenitors from which new neurons arise, were found in the brains of adult mammals, and discovered to undergo expansion in their populations when prompted by signaling molecules called growth factors and morphogens (11). The multiplication and differentiation of neural stem cells, which are residents of the central nervous system, is essential for neurogenesis (14). Neural stem cells are capable of generating all of the cell types of the nervous system, including astrocytes, glial cells, and what are called oligodendrocytes in the central nervous system and Schwann cells in the peripheral nervous system (11). Researchers Colucci-D’Amato and Bonita in fact state that, “To date neural stem cells have been isolated from nearly all areas of the embryonic brain and in a growing list of adult mammalian brain areas, including cerebellum and cortex” (11, p. 268).
Other advances, such as confocal microscopy and the identification of cellular markers which allowed the phenotype of cells to be characterized all culminated in the realization that neurogenesis occurs continuously in some brain area, such as the hippocampus and subventricolar zone (SVZ), the former of which is responsible for the formation and consolidation of memories (11). To date, neurogenesis has been shown to be influenced by various chemical, pharmacological, and environmental stimuli. For instance, work by researcher Fernando Nottebohm demonstrated the spontaneous replacement of neurons in the adult avian brain (15). In song birds such as canaries, which experience seasonal modification in their songs, new neurons are recruited into their neuronal circuitry in a way that may be dependent upon social and reproductive interactions, territorial defense, migratory patterns and food caching (15).
This all should serve as a beacon of hope for patients experiencing the ravages of neurodegenerative disease, as it may mean that epigenetics, or the way gene expression changes based on lifestyle factors, may lend itself to neurogenesis and the reversal of these scourges of mankind. For example, researchers state that an enriched environment, learning, exercise, exposure to different odorant molecules, and drugs such as antidepressants, steroids, and alcohol can all favorably or unfavorably impact neurogenesis (11). These newfound revelations are being used in fact as an impetus to find cures for a laundry list of neurodegenerative diseases (11).
Novel Therapy Shown to Grow New Nerve Cells
Despite this research, the prevailing view of neurodegenerative diseases such as Alzheimer’s and Parkinson’s is that their underlying pathophysiology, a relentless progression of neuronal death, remains irreversible (10). Thus far, then, approaches have aimed to slow or stop neuronal cell death or to develop disease-modifying treatments that could stabilize the rate of neurodegeneration (10). One non-pharmacological therapy that may be able to actually regenerate brain cells, however, is light in the near infrared range, also known as low-level laser or light emitting diode (LED) therapy that utilizes wavelengths in the red to infrared spectrum.
Near infrared light therapy has the potential to “mitigate ubiquitous processes relating to cell damage and death,” and may have applications in conditions that “converge on common pathways of inflammation and oxidative stress” (10). This is demonstrated by the widespread efficacy of near infrared light therapy in improving conditions including traumatic brain injury, ischemic stroke, major depression, and age-related macular degeneration (10). In traumatic brain injury, for example, treatment with near infrared light improves social, interpersonal, and occupational functions, reduces symptoms of post-traumatic stress disorder (PTSD), and is helpful for sleep (16).
Because near infrared light treatment improves cognitive and emotional dimensions (17) and enhances short-term memory and measures of sustained attention (18), researchers have long suspected its potential for neuropsychological disorders. In a revolutionary publication, scientists propose that infrared light is superior to pharmacological standard of care for these debilitating conditions given its neuron-saving abilities (10).
For instance, in mouse models of traumatic brain injury, near infrared light increases levels of brain-derived neurotrophic factor (BDNF), a protein which helps dying nerve cells survive (19). In addition, infrared light both improves neurological performance and increases the numbers of neuroprogenitor cells, the precursors to new neurons, in areas of the brain such as the dentate gyrus of the hippocampus and the sub ventricular zone (20).
Near Infrared Light Therapy in Alzheimer’s and Parkinson’s
Although human trials have not been yet conducted in Alzheimer’s disease, mouse studies show that near infrared treatment reduces its characteristic proteinopathies, decreasing brain levels of β-amyloid plaques and neurofibrillary tangles of tau proteins, while also ameliorating cognitive deficits (10). Cellular energy production, as indicated by levels of ATP, were increased in these studies alongside bolstered mitochondrial function and (10). In transgenic mouse models of Alzheimer’s, application of non-thermal near infrared light reversed significant deficits in working memory and significantly improved cognitive performance (21).
In animal models of Parkinson’s, near infrared treatment has been shown to rescue dopaminergic neurons, the subset that degenerate in this condition, from death (10). In addition, near infrared light treatment corrects the abnormal firing activity of neurons in deep subthalamic brain regions that occurs in parkinsonian conditions (22). Various animal models of Parkinson’s disease shown improved motor control and locomotor activity, as measured by both mobility and velocity, after near infrared is applied (10).
In a macaque monkey model of Parkinson’s, an optical fiber device that administered near infrared to the midbrain largely prevented the development of clinical signs of Parkinson’s when the animals were injected with a chemical known to induce this disorder (23). It also preserved a greater number of dopaminergic nigral cells compared to the monkeys that had not received infrared treatment (23). Limited case reports in humans have shown that near infrared administered through an intranasal apparatus improves symptoms in the majority of Parkinson’s patients, and that its application to the back of the head and upper neck reduced signs of Parkinson’s in one patient (10). Other reports indicate that gait, speech, cognitive function, and freezing episodes were improved in late-stage Parkinson’s patients who undertook this therapy (24), but the study was low-quality (10).
Mechanism of Action: How Near Infrared Promotes Neurogenesis
The ways in which near infrared promotes neurogenesis are multi-fold. There is evidence that near infrared light exerts a hormetic effect, acting as an adaptive or positive stressor. Another example of a hormetic effect is that exhibited by phytonutrients in fruits and vegetables, which act as antioxidants by paradoxically stimulating oxidative damage via a pro-oxidant mechanism. This in turn up-regulates our endogenous antioxidant defense system. Similarly, near infrared light activates cellular stress response systems by targeting a key enzyme in the electron transport chain which is responsible for mitochondrial-based energy production called cytochrome c oxidase, an enzyme that is fundamental to the cellular bioenergetics of nerve cells (25).
By accepting light in the near infrared range of the electromagnetic spectrum, this enzyme induces a change in the electrochemical potential of the mitochondrial membrane, jump-starting production of the cellular energy currency called adenosine triphosphate (ATP) and causing a mild burst in the synthesis of reactive oxygen species (ROS) (10). As a result, downstream signaling pathways are triggered which induce reparative and neuroprotective mechanisms, including neurogenesis, the creation of new synapses, and brain-based antioxidant and metabolic effects (25).
Restoration of mitochondrial function in the endothelial cells lining cerebral blood vessels may also help neurons survive by repairing the blood-brain barrier and vascular network which is compromised in neurogenerative conditions (10). Impressively, “This modulation of multiple molecular systems appears capable of both conditioning neurons to resist future damage and accelerating repair of neurons damaged by a previous or continuing insult” (10).
On the other hand, the application of near infrared light has been shown to elicit systemic effects, possibly via circulating molecular factors (10). In other words, light in the near infrared spectrum applied to a local area elicits benefits in distal tissues remote from the initial site, perhaps by stimulating immune cells that have a neuroprotective role (10). Another way in which near infrared light activates global effects in the body is by up-regulating the production of signaling molecules known as anti-inflammatory cytokines, while down-regulating pro-inflammatory cytokines (26).
Near infrared also mobilizes tissue repair processes by improving the migration of white blood cells to wounds, increasing neovascularization, or the formation of new blood vessels, and facilitating formation of collagen (27). There is also evidence that near-infrared light exposure causes stem cells from the bone marrow to navigate to the site of damage and to release so-called trophic factors such as BDNF, which enhances nerve cell function and survival (28). Lastly, a system of communication between the mitochondria in the brain and the mitochondria in the tissues may be at play, so that application of near infrared light at a point in the body far from the brain can lead to neural regeneration (10).
Practical Application of Near Infrared Light Therapy
The key to mitigating the burden of chronic illness lies in physiological regeneration, which is emerging as a physiological inevitability, even in regions of the body where it was previously not thought possible. The ability to regenerate, secondary to normal biological processes of cellular erosion and decay, is programmed into our body in order for us to regain homeostasis.
So-called “photobiomodulation,” which includes near infrared light therapy, has limitless possible applications, and has even been shown to improve animal models of wound healing, heart attack, spinal cord injury, stroke, arthritis, familial amylotropic lateral sclerosis (FALS), diabetic ulcers, carpal tunnel syndrome, major depression, generalized anxiety disorder, frontotemporal dementia (29) and traumatic brain injury (27).
The biggest obstacle with infrared light therapy in neurodegenerative disease is targeting the zone of pathology, “when there are many intervening body tissues, namely skin, thick cranium, and meninges, and brain parenchyma,” since there is considerable dissipation of the signal across each millimeter of brain tissue (10). This is less problematic in Alzheimer’s, where the target regions are more superficial structures, but less easily rectified in the case of Parkinson’s, where there is significant distance from cranium to the brainstem where neurodegeneration takes place (10).
With Alzheimer’s, optimal delivery would be a near infrared light-emitting helmet worn over the entire cranium (10). Parkinson’s patients can achieve symptomatic relief when near infrared is applied in this fashion, as this would influence the abnormal neural circuitry in the cortex. However, to circumvent the problem of the sheer distance to the region of pathology in the brainstem, researchers propose that the minimally invasive surgical implantation of an optical fiber device near the brain parenchyma would be ideal, which would deliver therapeutic levels of near infrared (10). Until these options are commercially available, photobiomodulation devices or near infrared saunas may be a viable option, although human studies have not proved their efficacy.
Given its large margin of safety and lack of adverse effects, near infrared light therapy should be offered as an option for patients suffering from a myriad of chronic conditions, but is especially promising for neurodegenerative diseases including Alzheimer’s and Parkinson’s and may even have future use in multiple sclerosis. Near infrared therapy is superior to the mainstay drug treatments for these diseases since pre-clinical studies have demonstrated proof-of-concept that near infrared either arrests or slows the underlying pathology of these disease processes, and leads to the birth of new neurons, rather than merely mitigating symptoms (10).
2. Goedert, M. (2015). Alzheimer’s and Parkinson’s diseases: the prion concept in relation to assembled Aβ, tau, and α-synuclein. Science, 349, 1255555.
3. Stone, J. (2008). What initiates the formation of senile plaques? The origin of Alzheimer-like dementias in capillary haemorrhages. Medical Hypotheses, 71, 347–359.
4. Gonzalez-Lima, F., Barksdale B.R., & Rojas J.C. (2014). Mitochondrial respiration as a target for neuroprotection and cognitive enhancement. Biochemical Pharmacology, 88, 584–593. 10.1016/j.bcp.2013.11.010
5. Bergman, H., & Deuschl, G. (2002). Pathophysiology of Parkinson’s disease: from clinical neurology to basic neuroscience and back. Movement Disorders, 7(Suppl. 3), S28–S40.
6. Lanciego, J.L., Luquin, N., & Obeso, J.A. (2012). Functional Neuroanatomy of the Basal Ganglia. Cold Springs Harbor Perspectives in Medicine, 2(12), a009621.
7. De Virgilio, A. et al. (2016). Parkinson’s disease: Autoimmunity and neuroinflammation. Autoimmunity Reviews, 15(10), 1005-1011. doi: 10.1016/j.autrev.2016.07.022.
8. Gitler A.D. et al. (2009). Alpha-synuclein is part of a diverse and highly conserved interaction network that includes PARK9 and manganese toxicity. Natural Genetics, 41, 308–315.
9. Exner, N. et al. (2012). Mitochondrial dysfunction in Parkinson’s disease: molecular mechanisms and pathophysiological consequences. EMBO Journal, 31, 3038–3062. 10.1038/emboj.2012.170
10. Johnstone, D.M. et al. (2015). Turning On Lights to Stop Neurodegeneration: The Potential of Near Infrared Light Therapy in Alzheimer’s and Parkinson’s Disease. Frontiers in Neuroscience, 9, 500. doi: 10.3389/fnins.2015.00500
11. Colucci-D’Amato, L., & Bonavita, V. (2006). The end of the central dogma of neurobiology: stem cells and neurogenesis in adult CNS. Neurological Science, 27(4), 266-270.
12. Altman, J. (1962). Are new neurons formed in the brains of adult mammals? Science, 135, 1127-1128.
13. Kaplan, M.S., & Hinds, J.W. (1977). Neurogenesis in the adult rat: electron microscopic analysis of light radioautographs. Science, 197, 1092-1094.
14. Martino, G. et al. (2011). Brain regeneration in physiology and pathology: the immune signature driving therapeutic plasticity of neural stem cells. Physiological Reviews, 91(4), 1281-1304.
15. Nottebohm, F. (2002). Why are some neurons replaced in adult brain? Journal of Neuroscience, 22(3), 624-628.
16. Naeser, M.A. et al. (2014). Significant improvements in cognitive performance post-transcranial, red/near-infrared light-emitting diode treatments in chronic, mild traumatic brain injury: open-protocol study. Journal of Neurotrauma, 31,(11), 1008-1017. doi: 10.1089/neu.2013.3244.
17. Barrett, D.W., & Gonzalez-Lima, F. (2013). Transcranial infrared laser stimulation produces beneficial cognitive and emotional effects in humans. Neuroscience, 230, 13-23. doi: 10.1016/j.neuroscience.2012.11.016.
18. Blanco, N.J., Maddox, W.T., & Gonzalez-Lima, F. (2015). Journal of Neuropsychology, 11(1),14-25. doi: 10.1111/jnp.12074.
19. Xuan, W. et al. (2013). Transcranial low-level laser therapy improves neurological performance in traumatic brain injury in mice: effect of treatment repetition regimen. PLoS ONE, 8, e53454.
20. Xuan, W. et al. (2014). Transcranial low-level laser therapy enhances learning, memory, and neuroprogenitor cells after traumatic brain injury in mice. Journal of Biomedical Optics, 191(10), 108003.
21. Michalikova, S. et al. (2008). Emotional responses and memory performance of middle-aged CD1 mice in a 3D maze: effects of low infrared light. Neurobiology of Learning and Memory, 89(4), 480-488.
22. Shaw, V.E. et al. (2012). Patterns of Cell Activity in the Subthalamic Region Associated with the Neuroprotective Action of Near-Infrared Light Treatment in MPTP-Treated Mice. Parkinsonian Disease, 2012, 29875. doi: 10.1155/2012/296875.
23. Darlot, F. et al. (2016). Near-infrared light is neuroprotective in a monkey model of Parkinson disease. Annals of Neurology, 79(1), 59-65. doi: 10.1002/ana.24542.
24. Maloney, R., Shanks, S., & Maloney J. (2010). The application of low-level laser therapy for the symptomatic care of late stage Parkinson’s disease: a non-controlled, non-randomized study. American Society of Laser Medicine and Surgery, 185.
25. Rojas, J.C., & Gonzalez-Lima, F. (2011). Low-level light therapy of the eye and brain. Eye and Brain, 3, 49–67.
26. Muili, K.A. et al. (2012). Amelioration of experimental autoimmune encephalomyelitis in C57BL/6 mice by photobiomodulation induced by 670 nm light. PLoS ONE, 7, e30655.
27. Chung, H. et al. (2012). The Nuts and Bolts of Low-level Laser (Light) Therapy. Annals of Biomedical Engineering, 40(2), 516-533.gma
28. Hou, S.T. et al. (2008). Permissive and Repulsive Cues and Signalling Pathways of Axonal Outgrowth and Regeneration. International Review of Cell and Molecular Biology, 267, 121-181.
29. Purushothuman, S. et al. (2013). The impact of near-infrared light on dopaminergic cell survival in a transgenic mouse model of parkinsonism. Brain Research, 1535, 61–70.
Ali Le Vere holds dual Bachelor of Science degrees in Human Biology and Psychology, minors in Health Promotion and in Bioethics, Humanities, and Society, and is a Master of Science in Human Nutrition and Functional Medicine candidate. Having contended with chronic illness, her mission is to educate the public about the transformative potential of therapeutic nutrition and to disseminate information on evidence-based, empirically rooted holistic healing modalities. Read more at @empoweredautoimmune on Instagram and atwww.EmpoweredAutoimmune.com: Science-based natural remedies for autoimmune disease, dysautonomia, Lyme disease, and other chronic, inflammatory illnesses.
Disclaimer: This article is not intended to provide medical advice, diagnosis or treatment. Views expressed here do not necessarily reflect those of GreenMedInfo or its staff.
More than five million Americans have Alzheimer’s disease, the most common type of dementia. Meanwhile, 740,000 people die annually in the U.S. because of heart disease and stroke. To resolve this health concern, scientists are studying the link between high levels of the amino acid homocysteine and these life-threatening conditions.
Research on homocysteine and health conditions
According to a study by researchers from the Lewis Katz School of Medicineat Temple University, B-complex vitamins play a crucial role when it comes to controlling homocysteine. Findings from the study, which was published in Molecular Psychiatry, determined that elevated levels of the amino acid can result in Alzheimer’s, along with other forms of dementia.
Alarmingly enough, there are many reports of vitamin B deficiencies in the U.S. More patients are also being diagnosed with Alzheimer’s disease. These concerns highlight the need to maintain healthy levels of these B-complex vitamins for disease prevention.
For the study, the researchers gave mice a diet lacking in vitamin B6, vitamin B9 (folate), and vitamin B12. Eight months after the mice were fed the B-complex deficient diet, the research team used a water maze test to gauge their memory and learning. Unlike the control mice that were fed a normal diet, the vitamin B-deficient mice had trouble learning a new task. The vitamin deficiency also affected their ability to remember it.
The study revealed that the mice brains had both elevated levels of homocysteine and “tau,” a protein that damages or destroys brain nerve cells/neurons. Tau can also disrupt synapses, which are the junctions that allow neuronal communication.
Mother Nature’s micronutrient secret: Organic Broccoli Sprout Capsules now available, delivering 280mg of high-density nutrition, including the extraordinary “sulforaphane” and “glucosinolate” nutrients found only in cruciferous healing foods. Every lot laboratory tested. See availability here.
Rodents deprived of B vitamins also had a 50 percent increase in neurofibrillary tau tangles in the hippocampus and cortex, two areas necessary for learning and memory. A hallmark of Alzheimer’s disease, tau tangles are also the main factors of “cell death, dementia, and neurodegenerative conditions.”
During the study, the research team discovered data linked to the formation of tau tangles. The authors shared that one of the key changes caused by high levels of homocysteine was elevated levels of 5-lipoxygenase (5LO), the pro-inflammatory chemical that causes tau tangles.
Further studies can help determine if thwarting the production of 5LO can prevent or reverse the brain damage linked to elevated homocysteine levels.
Domenico Pratico, study leader and a professor in the Departments of Pharmacology and Microbiology at Lewis Katz, commented that high homocysteine levels were previously associated with amyloid beta plaques that are also linked to Alzheimer’s disease.
However, until the study findings from the Lewis Katz study, the connection with tau tangles remained unknown.
How can you prevent hyperhomocysteinemia?
Homocysteine is a non-protein amino acid that can naturally be found in the human body. The amino acid is a byproduct of the metabolism of another amino acid called methionine.
Hyperhomocysteinemia, the medical term for high homocysteine levels, may have a genetic component. The disease can also occur due to deficiencies of B vitamins and folic acid, improper diet, or stress.
Data from the study showed that elevated homocysteine damages fragile arterial linings, causes inflammation and oxidative stress, and minimizes blood flow to the heart and brain. These destructive processes can eventually cause atherosclerosis and coronary artery disease. In fact, blood levels of homocysteine can be used to correctly predict the risk of heart disease.
Research has also revealed that hyperhomocysteinemia is linked to a whopping 42 percent increase in the risk of the narrowing of the carotid arteries. Individuals with elevated homocysteine who have already had a heart attack are at a 30 percent higher risk of suffering another adverse event such as another heart attack, stroke, or even death.
High homocysteine levels can also double your chance of developing dementia. Consult a healthcare professional if you want to check your homocysteine levels with a simple blood test. Levels below 10 micromoles per liter (umol/L) are healthy while seven umol/L to eigh umol/L is optimal.
The following tips can help prevent a vitamin B-complex deficiency and hyperhomocysteinemia:
Eat foods rich in B12 like grass-fed beef, organic dairy, and wild-caught salmon.
Take B complex vitamin supplements, especially if you already have hyperhomocysteinemia. Talk to a healthcare professional before you take any supplements.
To lower high homocysteine levels, take 25 to 100 milligrams (mg) of vitamin B2, 100 to 200 mg of vitamin B6, 1,000 to 10,000 micrograms (mcg) of vitamin B9, and 300 to 1,000 mcg of vitamin B1 (methylcobalamin) daily. These B complex vitamins, which can all help detoxify homocysteine, must be taken together with the omega-3 fatty acids eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) in fish oil.
To prevent hyperhomocysteinemia and lower the risk of developing life-threatening health conditions, follow a healthy diet and increase your intake of B complex vitamins.
You can read more articles about how to manage or prevent Alzheimer’s and lower your homocysteine levels at Alzheimers.news.
(Natural News) Cannabis may still be prohibited by the U.S. federal government, but that doesn’t mean the prized plant isn’t a medicine. While federal prohibition may make scientific research on cannabis harder to complete, many studies have shown that the plant has wide-reaching medicinal benefits. Research continues to show that for dozens of conditions, cannabis could be a natural solution.
As of 2018, 29 U.S. states have legalized cannabis for medical use. In the U.S., legalized medical cannabis is gaining more support than ever, with an 84-percent approval rating according to recent survey data.
Even the National Institutes of Health recognizes the medical benefits of cannabis – and it’s a federal organization. All this has left many people wondering why cannabis prohibition is still running strong in the U.S. But despite prohibition’s dampening effect on research, the science on medicinal cannabis continues to pour in. Here are 10 health conditions scientifically shown to have success with cannabis treatment:
1. Cannabis can relieve many types of pain
For chronic pain sufferers of any kind, cannabis can bring relief with fewer side effects and less risk than many mainstream treatments. Studies of patients with peripheral neuropathy have shown that pain reduction can be observed within a week of regular use. CBD, or cannabidiol, is thought to be the key pain alleviator here; reports say that it “reduces the inflammatory response and binds to TRPV1 receptors, which are capable of mediating antihyperalgesic effects.”
100% organic essential oil sets now available�for your home and personal care, including Rosemary, Oregano, Eucalyptus, Tea Tree, Clary Sage and more, all 100% organic and laboratory tested for safety. A multitude of uses, from stress reduction to topical first aid. See the complete listing here, and help support this news site.�
Chronic pain can come from many different sources, but for fibromyalgia patients, cannabis could be very effective. Studies also show that it can relieve other types of pain, like arthritis. While not every person with chronic pain finds relief from cannabis, many do. In fact, pain is the most common reason people ask their doctors for medical marijuana.
2. Relief from anxiety
In the right amount, cannabis has been shown to reduce anxiety and stress in users. However, the dose makes the poison: For many people, too much cannabis can cause anxiety to spike instead. CBD oil can also help reduce feelings of anxiousness.
3. Eases depression
Studies have shown that the infamous plant has anti-depressive benefits as well. Specifically, the plant compound CBD has been shown to exhibit similar effects on depression as pharmaceutical antidepressants.
4. Cannabis protects brain cells
Despite popular belief, cannabinoids found in the cannabis plant can actually help protect your brain – especially CBD. Sources say that CBD is “proven to reduce short-term brain damage and was associated with extracerebral benefits.”
5. Fights Alzheimer’s disease
In addition to protecting the brain against damage, studies have shown that THC, a compound in cannabis, can slow down the progression of Alzheimer’s disease. In fact, the plant may help reverse the degenerative condition entirely.
6. Relieves the symptoms of MS
Multiple sclerosis (MS) is a serious disease with no currently known cure. But studies have shown that cannabis can provide much needed relief to people who have MS – especially those struggling with painful muscle spasms. Muscle spasticity effects an estimated 90 percent of MS patients. Studies show that cannabis can help relieve this symptom; survey data indicates that as many as 97 percent of MS patients could benefit from it.
7. Eliminates nausea, increases hunger
Cannabis can help reduce feelings of nausea in patients suffering from cancer, HIV/AIDS and other debilitating conditions. Furthermore, in patients with poor appetite, the plant medicine may help increase overall tolerance of food.
8. Treats IBD
For patients with inflammatory bowel diseases (IBD), cannabis could be a novel treatment that provides symptom relief. Studies suggest that both THC and CBD (two well-known cannabinoids) play an important role in gut function – and the benefits can’t be ignored.
9. Cannabis reduces seizure activity
There is much to support the fact that cannabis reduces seizure activity in people with seizure disorders. There is a mountain of evidence which shows children with intractable seizures have experienced a drastic reduction in seizure frequency thanks to cannabis.
THE CLEAN CYCLE Lack of sleep may contribute to Alzheimer’s disease by robbing the brain of the time it needs to wash away sticky proteins.
Neuroscientist Barbara Bendlin studies the brain as Alzheimer’s disease develops. When she goes home, she tries to leave her work in the lab. But one recent research project has crossed into her personal life: She now takes sleep much more seriously.
Bendlin works at the University of Wisconsin–Madison, home to the Wisconsin Registry for Alzheimer’s Prevention, a study of more than 1,500 people who were ages 40 to 65 when they signed up. Members of the registry did not have symptoms of dementia when they volunteered, but more than 70 percent had a family history of Alzheimer’s disease.
Since 2001, participants have been tested regularly for memory loss and other signs of the disease, such as the presence of amyloid-beta, a protein fragment that can clump into sticky plaques in the brain. Those plaques are a hallmark of Alzheimer’s, the most common form of dementia.
Each person also fills out lengthy questionnaires about their lives in the hopes that one day the information will offer clues to the disease. Among the inquiries: How tired are you?
Some answers to the sleep questions have been eye-opening. Bendlin and her colleagues identified 98 people from the registry who recorded their sleep quality and had brain scans. Those who slept badly — measured by such things as being tired during the day — tended to have more A-beta plaques visible on brain imaging, the researchers reported in 2015 in Neurobiology of Aging.
In a different subgroup of 101 people willing to have a spinal tap, poor sleep was associated with biological markers of Alzheimer’s in the spinal fluid, Bendlin’s team reported last year in Neurology. The markers included some related to A-beta plaques, as well as inflammation and the protein tau, which appears in higher levels in the brains of people with Alzheimer’s.
Bendlin’s studies are part of a modest but growing body of research suggesting that a sleep-deprived brain might be more vulnerable to Alzheimer’s disease. In animal studies, levels of plaque-forming A-beta plummet during sleep. Other research suggests that a snoozing brain runs the “clean cycle” to remove the day’s metabolic debris — notably A-beta — an action that might protect against the disease. Even one sleepless night appears to leave behind an excess of the troublesome protein fragment (SN Online: 7/10/17).
But while the new research is compelling, plenty of gaps remain. There’s not enough evidence yet to know the degree to which sleep might make a difference in the disease, and study results are not consistent.
A 2017 analysis combined results of 27 studies that looked at the relationship between sleep and cognitive problems, including Alzheimer’s. Overall, poor sleepers appeared to have about a 68 percent higher risk of these disorders than those who were rested, researchers reported last year in Sleep. That said, most studies have a chicken-and-egg problem. Alzheimer’s is known to cause difficulty sleeping. If Alzheimer’s both affects sleep and is affected by it, which comes first?
For now, the direction and the strength of the cause-and-effect arrow remain unclear. But approximately one-third of U.S. adults are considered sleep deprived (getting less than seven hours of sleep a night) and Alzheimer’s is expected to strike almost 14 million U.S. adults by 2050 (5.7 million have the disease today). The research has the potential to make a big difference.
Dream weavers
It would be easier to understand sleep deprivation if scientists had a better handle on sleep itself. The brain appears to use sleep to consolidate and process memories (SN: 6/11/16, p. 15) and to catalog thoughts from the day. But that can’t be all. Even the simplest animals need to sleep. Flies and worms sleep.
But mammals appear to be particularly dependent on sleep — even if some, like elephants and giraffes, hardly nod off at all (SN: 4/1/17, p. 10). If rats are forced to stay awake, they die in about a month, sometimes within days.
And the bodies and brains of mice change when they are kept awake, says neurologist David Holtzman of Washington University School of Medicine in St. Louis. In one landmark experiment, Holtzman toyed with mice’s sleep right when the animals’ brain would normally begin to clear A-beta. Compared with well-rested mice, sleep-deprived animals developed more than two times as many amyloid plaques over about a month, Holtzman says.
Losing sleep
Alzheimer’s disease disrupts sleep. And disrupted sleep itself might encourage Alzheimer’s by allowing buildup of amyloid-beta, or A-beta, which is thought to lead to the death of neurons. This cycle of sleep deprivation can also affect levels of the hormone melatonin, which helps the body to sleep, and can interfere with metabolism, a disruption that is also a risk factor for Alzheimer’s.
Source: Y. Saeed and S.M. Abbott/Current Neurology and Neuroscience Reports 2017
He thinks Alzheimer’s disease is a kind of garbage collection problem. As nerve cells, or neurons, take care of business, they tend to leave their trash lying around. They throw away A-beta, which is a leftover remnant of a larger protein that is thought to form connections between neurons in the developing brain, but whose role in adults is still being studied. The body usually clears away A-beta.
But sometimes, especially when cheated on sleep, the brain doesn’t get the chance to mop up all the A-beta that the neurons produce, according to a developing consensus. A-beta starts to collect in the small seams between cells of the brain, like litter in the gutter. If A-beta piles up too much, it can accumulate into plaques that are thought to eventually lead to other problems such as inflammation and the buildup of tau, which appears to destroy neurons and lead to Alzheimer’s disease.
About a decade ago, Holtzman wanted to know if levels of A-beta in the fluid that bathes neurons fluctuated as mice ate, exercised, slept and otherwise did what mice do. It seemed like a run-of-the-mill question. To Holtzman’s surprise, time of day mattered — a lot. A-beta levels were highest when the animals were awake but fell when the mice were sleeping (SN: 10/24/09, p. 11).
“We just stumbled across this,” Holtzman says. Still, it wasn’t clear whether the difference was related to the hour, or to sleep itself. So Holtzman and colleagues designed an experiment in which they used a drug to force mice to stay awake or fall asleep. Sure enough, the A-beta levels in the brain-bathing fluid rose and fell with sleep, regardless of the time on the clock.
A-beta levels in deeply sleeping versus wide-awake mice differed by about 25 percent. That may not sound like a dramatic drop, but over the long term, “it definitely will influence the probability [that A-beta] will aggregate to form amyloid plaques,” Holtzman says.
The study turned conventional thinking on its head: Perhaps Alzheimer’s doesn’t just make it hard to sleep. Perhaps interrupted sleep drives the development of Alzheimer’s itself.
Published in Science in 2009, the paper triggered a flood of research into sleep and Alzheimer’s. While the initial experiment found that the condition worsens the longer animals are awake, research since then has found that the reverse is true, too, at least in flies and mice.
Using fruit flies genetically programmed to mimic the neurological damage of Alzheimer’s disease, a team led by researchers at Washington University School of Medicine reversed the cognitive problems of the disease by simply forcing the flies to sleep (SN: 5/16/15, p. 13).
Researchers from Germany and Israel reported in 2015 in Nature Neuroscience that slow-wave sleep — the deep sleep that occupies the brain most during a long snooze and is thought to be involved in memory storage — was disrupted in mice that had A-beta deposits in their brains. When the mice were given low doses of a sleep-inducing drug, the animals slept more soundly and improved their memory and ability to navigate a water maze.
Diagnosed with Alzheimer’s in 2015, 75-year-old Brenda Whittle still enjoys jigsaws, sewing and dancing. New activities are less appealing, but participating in Alzheimer’s research and drug trials is an exception. She’s so at ease with loud brain scans, she even falls asleep during them.
“This can’t be sustained by any medical health system – it is too much in terms of numbers, says Antonella Santuccione-Chadha, a physician and Alzheimer’s specialist based in Switzerland. “And as women are more confronted by the disease, we need to investigate the differences between the male and female specifics of it.”
Much of the gender gap comes down to one of dementia’s biggest risk factors: age. The older you are, the more likely you are to develop late-onset Alzheimer’s. Women typically live longer than men, so more have dementia.
The older you are, the more likely to develop dementia (Credit: Getty Images)
But recent research hints that we would be wrong to assume that ageing means Alzheimer’s is inevitable. Results from two major Cognitive Function and Ageing Studies (CFAS) suggest that over the last 20 years, new dementia cases in the UK have dropped by 20% – driven mostly by a fall in incidence among men over 65 years old.
Over the past 20 years, new dementia cases in the UK have dropped by 20% – driven mostly by a fall in incidence among men over 65
Experts say this may be because of public health campaigns targeting heart disease and smoking. Both are risk factors for Alzheimer’s. But because men tend to get heart disease younger and smoke more than women, these campaigns also may have helped stave off these risk factors more for men than women.
“Sex-specific prevention might start from having more of this information about female-specific risk factors,” says Maria Teresa Ferretti, a biomedical researcher in the field of Alzheimer’s disease at the University of Zurich.
The “quick facts” provided by the Alzheimer’s Association are pretty concerning: More than five million people in America are living with Alzheimer’s, and that number is projected to reach 16 million by the year 2050. As the sixth leading cause of death in our nation, it kills more Americans than prostate cancer and breast cancer combined. Someone in the U.S. develops Alzheimer’s every 66 seconds; will you be one of them?
With statistics like these, it’s no wonder that people want to do everything they can to reduce their odds. However, it’s also important to note that Alzheimer’s is only one of the potential causes of dementia. While many people use the terms interchangeably, Alzheimer’s is really only responsible for around 50 to 70 percent of dementia cases. The misleading terminology is obscuring one very dark fact about dementia: Many times, it’s being caused not by something scientists are still struggling to understand like Alzheimer’s but rather by things that are masquerading as tools for good health; vaccines and prescription drugs.
In fact, the Alzheimer’s Association that publicizes these statistics is subsidized by Big Pharma. It’s simply good business sense that they want people to believe that every memory-loss patient falls under the Alzheimer’s umbrella because then they can sell you drugs that purportedly address it. Their research has led them to an approach that pays dividends: promoting and destigmatizing what many think of as “mental illnesses,” making them seem unpreventable but manageable with drugs. Many people who work for the Alzheimer’s Association and similar organizations are well-meaning people who want to help and are often unaware of the connection to Big Pharma.
You have more control over “dementia” than you’re being led to believe
It’s no coincidence that dementia cases have been spiking during the same time that children and adults alike are being over-vaccinated (flu shot, anyone?) and the over-prescription of brain-altering drugs like antidepressants is prevalent.
A help guide based on a Harvard University report admits as much. According to the report, “medications are common culprits in mental decline.” As the body ages, the liver’s efficiency when it comes to metabolizing drugs declines, and the kidneys do not eliminate them as quickly as they once did. This causes the drugs to accumulate in the body, which means those who take multiple medications are particularly susceptible to this effect.
Included in the list of drugs published in the guide that cause dementia-like symptoms are antidepressants, anti-anxiety medications, sedatives, corticosteroids, narcotics, antihistamines, cardiovascular drugs, and anticonvulsants. It’s a very broad range of drugs, and many elderly people take medications from one or more of those categories. In fact, you might want to go check your medicine cabinet right now.
A study published in JAMA Internal Medicine correlated the use of popular medications like Benadryl and other anticholinergic drugs with dementia onset. According to the researchers, patients who took these medications for three years or more had a 54 percent higher chance of going on to develop the disorder.
Vaccines are also responsible for causing symptoms mistaken for dementia. People in their 40s are increasingly being diagnosed with “dementia,” and experts believe that environmental factors must be responsible in these cases. Mercury-containing thimerosalwas used widely in childhood vaccines until 2001 and remains in some vaccines, including flu shots, to this day. A study published in the Journal of Alzheimer’s Disease found that exposure to mercury could produce many of the changes that are seen in Alzheimer’s patients, including impaired cognitive function and memory as well as confusion.
Researcher Richard Deth stated: “Mercury is clearly contributing to neurological problems, whose rate is increasing in parallel with rising levels of mercury. It seems that the two are tied together.”
Another common ingredient found in vaccines, aluminum, has been linked to dementia as well.
It’s a pretty smart way to keep the profit machine turning for Big Pharma: Convince people they need vaccines or drugs, and when those vaccines or drugs cause further side effects and illnesses, sell them even more drugs to counteract them. And the best part for them is that because mental decline is involved, it reduces the chances that people will wake up to what is really going on here.
Even one night of lost sleep may cause the brain to fill with protein chunks that have long been linked to the development of Alzheimer’s disease, a new study warns.
People deprived of sleep for one night experience an immediate and significant increase in beta amyloid, a substance that clumps together between neurons to form plaques that hamper the brain‘s ability to function, researchers found.
“We certainly show that even one night of sleep deprivation can increase the levels of these harmful beta amyloid compounds,” said study author Ehsan Shokri-Kojori, a research fellow with the U.S. National Institute on Alcohol Abuse and Alcoholism.
“That’s a very logical assumption, I would say, and it’s consistent with prior research,” he said.
Previous mouse and human studies have found potential links between too little sleep and an accumulation of beta amyloid in the brain, researchers said in background notes. However, many of the human studies have relied on self-reports of sleep quality.
So Shokri-Kojori and his team decided to create an experiment that would more precisely test the effect of sleep deprivation on beta amyloid levels in humans.
They recruited 20 healthy people with no history of brain disorders, and had them spend two nights in the lab—one in which they were allowed to get a good night’s rest, and another in which they didn’t sleep a wink.
The morning after both nights, the participants underwent brain scans to assess their levels of beta amyloid.
The researchers found that sleep deprivation was associated with a significant increase in beta amyloid in the brain, when compared with a good night’s sleep.
Brain imaging after one night of sleep deprivation revealed beta-amyloid accumulation in the hippocampus and thalamus, regions affected by Alzheimer’s disease Credit: Proceedings of the National Academy of Sciences
Scientists have new evidence that suggests that THC inhibits the formation of amyloid plaques by blocking the enzyme in the brain that produces them. Neurodegenerative diseases such as Alzheimer’s are caused by the poor formation of those proteins in the brain.
An active compound in marijuana called tetrahydrocannabinol (THC) has been found to promote the removal of toxic clumps of amyloid beta protein in the brain, which are thought to kickstart the progression of Alzheimer’s disease.
The finding supports the results of previous studies that found evidence of the protective effects of cannabinoids, including THC, on patients with neurodegenerative disease.
“Although other studies have offered evidence that cannabinoids might be neuroprotective against the symptoms of Alzheimer’s, we believe our study is the first to demonstrate that cannabinoids affect both inflammation and amyloid beta accumulation in nerve cells,” says one of the team, David Schubert from the Salk Institute for Biological Studies in California.
Schubert and his colleagues tested the effects of THC on human neurons grown in the lab that mimic the effects of Alzheimer’s disease.
If you’re not familiar with this special little compound, it’s not only responsible for the majority of marijuana’s psychological effects – including the high – thanks to its natural pain-relieving properties, it’s also been touted as an effective treatment for the symptoms of everything from HIV and chemotherapy to chronic pain, post-traumatic stress disorder, and stroke.
In fact, THC appears to be such an amazing medical agent, researchers are working on breeding genetically modified yeast that can produce it way more efficiently than it would be to make synthetic versions.
The compound works by passing from the lungs to the bloodstream, where it attaches to two types of receptors, cannabinoid receptor (CB) 1 and 2, which are found on cell surfaces all over the body.
In the brain, these receptors are most concentrated in neurons associated with pleasure, memory, thinking, coordination and time perception, and usually bind with a class of lipid molecules called endocannabinoids that are produced by the body during physical activity to promote cell-to-cell signaling in the brain.
But THC can also bind to them in much the same way, and when they do, they start messing with your brain’s ability to communicate with itself. The can be a good and a bad thing, because while you might forget something important or suddenly be incapable of swinging a baseball bat, you’ll probably feel amazing, and want to eat all the snacks:
Over the years, research has suggested that by binding to these receptors, THC could be having another effect on aging brains, because it appears to helps the body clear out the toxic accumulations – or ‘plaques’ – of amyloid beta.
No one’s entirely sure what causes Alzheimer’s disease, but it’s thought to result from a build-up of two types of lesions: amyloid plaques and neurofibrillary tangles.
Amyloid plaques sit between the neurons as dense clusters of beta-amyloid molecules – a sticky type of protein that easily clumps together – and neurofibrillary tangles are caused by defective tau proteins that clump up into a thick, insoluble mass in the neurons.
It’s not clear why these lesions begin to appear in the brain, but studies have linked inflammation in the brain tissue to the proliferation of plaques and neurofibrillary tangles. So if we can find something that eases brain inflammation while at the same time encourages the body to clear out these lesions, we could be on the way to finding the first effective treatment for Alzheimer’s ever.
Back in 2006, researchers at the Scripps Research Institute found that THC inhibits the formation of amyloid plaques by blocking the enzyme in the brain that produces them, and now Schubert and his team have demonstrated that it can also eliminate a dangerous inflammatory response from the nerve cells, ensuring their survival.
“Inflammation within the brain is a major component of the damage associated with Alzheimer’s disease, but it has always been assumed that this response was coming from immune-like cells in the brain, not the nerve cells themselves,” says one of the team, Antonio Currais.
“When we were able to identify the molecular basis of the inflammatory response to amyloid beta, it became clear that THC-like compounds that the nerve cells make themselves may be involved in protecting the cells from dying.”
It’s exciting stuff, but it’s so far only been demonstrated in neurons in the lab, so the next step will be for Schubert and his team to observe the link between THC and reduced inflammation and plaque build-up in a clinical trial. And they’ve reportedly already found a drug candidate called J147 that appears to have the same effects as THC, so this might be the way they can test the effects of THC without the government getting in the way.
By Crawford KilianToday | TheTyee.ca Crawford Kilian is a contributing editor of The Tyee and at 77 considers himself “cognitively OK.”
Edith and Pat McGeer have investigated Alzheimer’s and other neurological problems for decades. Photo from McGeer and Associates.
According to the Alzheimer Society of Canada, 1,125,000 Canadians will have dementia in 20 years. The cumulative economic burden will be $872 billion, and the demand for long-term care will increase tenfold.
In 2008, just over 100,000 new dementia cases were diagnosed each year in Canada. By 2038, the ASC predicts more than 250,000 new cases each year. About half of those diagnosed with dementia will have Alzheimer’s disease.
Like death itself, such numbers don’t bear thinking about. Nor do we want to think about our own partners and children dedicating their old age to caring for us.
But a lot of experts are thinking hard about this problem, and one of them is 90-year-old neuroscientist Dr. Patrick McGeer. Now CEO of Aurin Biotech, McGeer had a long career at the University of British Columbia as well as a tumultuous one as a Liberal and Socred MLA and cabinet minister from 1962 to 1986. He may have dropped out of the public eye after leaving politics, but McGeer has kept very busy.
Since 1990, McGeer has been studying the causes of Alzheimer’s. Now, almost 30 years later, he says a simple test can identify those at risk, and an over-the-counter pain reliever can prevent it.
When we hear a strong claim, we expect strong evidence. And if the evidence holds up, we need to explore the implications.
A ‘crazy hypothesis’
McGeer’s evidence is indeed generally strong. In a recent review article in the Journal of Alzheimer’s Disease, McGeer and his colleagues summarize how their view of Alzheimer’s went from a “crazy hypothesis” in the 1980s to today’s widely accepted view that neuroinflammation causes damage to brain cells. That in turn results in Alzheimer’s.
In 1990, McGeer reported in the British journal The Lancet that patients using nonsteroidal anti-inflammatory drugs (NSAIDS) for rheumatoid arthritis seemed to develop fewer cases of Alzheimer’s than statistics said they should. Further research indicated that a major cause of neuroinflammation was a buildup of “amyloid-β” protein in the brain. Eventually, over years, this Aβ buildup destroys nearby nerve cells and results in Alzheimer’s.
Further research by McGeer and others established that NSAIDS — like aspirin and ibuprofen — could reduce the number of Alzheimer’s cases below the rate to be expected in a given group. But there was no effect unless the NSAIDS were administered well before the members of the group were statistically likely to be diagnosed with Alzheimer’s. Once diagnosed, Alzheimer’s wasn’t affected by NSAIDS.
McGeer and his team found a solution: Aβ may accumulate in brain cells, but it’s produced in all body tissues. A simple saliva test seems able to detect Aβ levels — and these can predict the likelihood that a given individual will develop Alzheimer’s. Some people produce low levels of Aβ all their lives, and don’t develop Alzheimer’s; other produce high levels, and are virtually certain to develop it.
Six phases of Alzheimer’s
In their review article, McGeer and his colleagues wrote: “A theoretical construct suggests the development of AD [Alzheimer’s] goes through six phases, each with decreasing opportunity for therapeutic intervention. Since the prevalence of clinical AD commences at age 65, the prevalence for actual AD disease onset can be hypothesized to occur at least 10 years earlier, or at age 55. Without intervention, the prevalence will then double every five years…. Any strategy which limits Aβ production, enhances its clearance, or prevents its aggregation, should be disease modifying. Effectiveness of treatment should be measurable by CSF Aβ levels returning toward normal.”
Dr. Mary Newport, desperate after orthodox medical attempts failed her husband’s dwindling dysfunction from Alzheimer’s, discovered coconut oil and rescued him from having to be put away in a special home. His remarkable recovery went viral on the internet and Dr. Newport wrote a 2011-12 bestseller, Alzheimer’s Disease: What if There Was a Cure?
Both the Internet’s coverage and her book started an international grassroots movement of folks with very early dementia, even just brain fog and senior moments, trying themselves or helping relatives diagnosed with Alzheimer’s. Thus creating a wave of success reports and testimonial videos.
However successful those reports were, mainstream medicine wasn’t recognizing coconut oil as an efficacious aid for Alzheimer’s or any type of dementia. Even the Alzheimer’s Association and other similar groups still won’t recommend or mention coconut oil to Alzheimer’s sufferers simply because there are no peer reviewed studies.
But somehow, a recent peer reviewed study has been completed and published with another in progress with positive results.
COCONUT OIL: NON-ALTERNATIVE DRUG TREATMENT AGAINST ALZHEIMER´S DISEASE (published December 1, 2015 by Nutricion Hospitalaria)
This clinical trial was conducted in Spain, which explains why the study was done to begin with. Spain and other EU nations and middle eastern nations, including Israel, seem less restricted with what they can study without pharmaceutical funding. Spain and Israel have done considerable research and human studies on cannabis’ medical applications, for example.
The Spanish clinical trial involved following Alzheimer’s patients of varying ages and genders with and without diabetes diagnoses to determine if coconut oil consumption had any effect on reducing their Alzheimer’s mental dysfunction.
They used cognitive testing before and after the clinical trial to determine changes. The intervention group was fed 40 ml of coconut oil daily, which comes to 2.7 tablespoons. Three tablespoons or more daily is recommended by experts, depending on dementia severity and coconut oil digestive tolerance.
The researchers noted “a statistically significant increase in test score … and therefore an improvement in cognitive status, improving especially women’s, those without diabetes mellitus type II, and severe patients.”
The Spanish researchers concluded, “this study, although preliminary, demonstrated the positive influence of coconut oil at the cognitive level of patients with Alzheimer’s, this improvement being dependent on sex, presence or absence of diabetes and degree of dementia.” (Source)
Another clinical study, supported by private funding of course, on the effects of coconut oil for Alzheimer’s has been underway in Florida. It was started in 2013 at the University of South Florida’s Health Byrd Alzheimer’s Institute, located in Tampa, Florida.
Publishing for that study is expected in 2016. But word on the street is that there has been a high success rate.
The Fallacy of No and Low Fat Dieting
First, let’s clear the air on the outdated coconut oil saturated fat issue. Clean saturated fats that are not hydrogenated or otherwise adulterated are necessary for overall and brain health. They are not the source of obesity and heart disease.
The notoriously faulty Ancel Keys Seven Countries Study, which was accepted whole cloth by the medical establishment, media, and the processed cooking oil and margarine manufacturers in the 1950s. It gave Crisco and others the platform to pitch their unhealthy processed hydrogenated oils containing trans-fats to replace coconut oil, palm oil, and butter.
It’s been determined that hydrogenated and semi-hydrogenated oils containing trans-fats are more responsible for obesity, heart disease, and various autoimmune diseases including rheumatoid arthritis.
Refined carbohydrates, added high sugar, especially high fructose corn syrup (HFCS), have added to the rise in obesity with heart problems remaining at the to of disease morbidity despite the no or low fat mania of several decades.
Pharmaceutical Failures for Alzheimer’s
So far, not one pharmaceutical drug designed for Alzheimer’s has been successful with anything but creating adverse side effects. That’s why Alzheimer’s is considered “untreatable”. Big Pharma has nothing to offer so nothing exists from the AMA perspective.
One study discovered that many with high cholesterol counts live to well into their 80s. Ironically, blockbuster Big Pharma revenue statin drugs cause more by doing what they claim, reducing cholesterol counts.
Coconut Oil for Alzheimer’s Prevention and Reversal
From Dr. Stephanie Steneff: The brain represents only 2% of the body’s total mass, but contains 25% of the total cholesterol. Cholesterol is required everywhere in the brain as an antioxidant, an electrical insulator (in order to prevent ion leakage), as a structural scaffold for the neural network, and a functional component of all membranes. Cholesterol is also utilized in the wrapping and synaptic delivery of the neurotransmitters. It also plays an important role in the formation and functioning of synapses in the brain.
Coconut oil differs from most other saturated fats, essentially fats that remain solid at room temperature, because it contains medium chain triglycerides or MCTs. Unlike the more common long chain triglycerides (LCTs), MCTs are easily digested by the liver and converted to energy in the form of ketones.
Long chain triglycerides or fatty acids don’t get readily converted into ketones that can be used by cells for energy. Much of LCTs or LCFAs wind up getting stored as fat.
Brain cells that have difficulty utilizing insulin for glucose wind up starving to death. Whole sections of the brain become useless as from the lack of energy caused by decreased glucose utilization.
This similarity to diabetes 2 with it’s cellular inability to process insulin, or insulin resistance, that is a major reason why Alzheimer’s or any similar dementia is considered diabetes 3.
The solution for diabetes 3 is ketones derived from the liver’s processing coconut oil MCTs. The ketones furnish brain cells with what they need regardless of insulin resistance that hampers glucose cellular metabolism. Ketones also increase blood circulation in the brain, a nice byproduct.
But ketones are easy come easy go. One needs to maintain a steady diet of healthy virgin coconut oil at the recommended pace of three or more tablespoons daily to maintain the cells energy levels.
The building blocks of brain matter are made from cholesterol. Coconut oil supplies that as well as the ketones that provide the brain energy despite insulin-glucose issues. With coconut oil, one gets the benefits of better brain structure and increased energy capacity.
Paul Fassais a contributing staff writer for REALfarmacy.com. His pet peeves are the Medical Mafia’s control over health and the food industry and government regulatory agencies’ corruption. Paul’s contributions to the health movement and global paradigm shift are well received by truth seekers. Visit his blog by following this link and follow him on Twitter here
Scientists have been aware of aluminum’s neurotoxicity for decades.
Although aluminum’s apologists have tried to shroud the metal’s risks in manufactured controversy, a growing number of reports by researchers in the United Kingdom, France, Canada, Israel, the U.S. and elsewhere has furnished substantive evidence linking aluminum to neuropathology, including the epidemics of Alzheimer’s disease (AD) and autism spectrum disorder (ASD).
Aluminum levels were particularly high in the male brains, including in a 15-year-old boy with ASD who had the study’s single highest brain aluminum measurement.
Dr. Christopher Exley — one of the world’s leading experts on aluminum toxicity — has shown that chronic intoxication with myriad forms of this “ubiquitous and omnipresent metal” is exacting a high price on human health.
Dr. Exley and other aluminum experts such as molecular biologist Dr. Lucija Tomljenovic have confirmed that aluminum readily and actively traverses the blood-brain barrier to selectively accumulate in brain tissues, where it induces unwelcome changes in brain biochemistry.
As Dr. Exley has noted, “There are no ‘normal’ levels of brain aluminum,” meaning that “its presence in brain tissue, at any level, could be construed as abnormal” [emphasis added].
Below is a video of him speaking on his study.
Documenting Aluminum in the ASD Brain
In light of the fact that even minute amounts of aluminum can have adverse neurological consequences, Dr. Exley’s newest paper — which reports on the first-ever study of aluminum in ASD brain tissue — is groundbreaking.
Published in the Journal of Trace Elements in Medicine and Biology, the paper documents some of the highest values for aluminum in human brain tissue ever recorded. Using a two-pronged study design (see box), the researchers measured and characterized aluminum deposits in brain tissues from five to ten ASD donors, most of whom died in their teens or twenties.
Study DesignQuantitative component: First, the investigators used graphite furnace atomic absorption spectrometry (GRAAS) to measure aluminum content in frozen brain tissue samples.
Frozen tissue was available from one female donor (age 44) and four male donors (ages 15, 22, 33 and 50) who, when alive, had a confirmed ASD diagnosis. The researchers quantified aluminum levels in 59 tissue samples representing five different areas of the brain (frontal, parietal, occipital, temporal and hippocampal).
Qualitative component: Using a technique called fluorescence microscopy, the researchers visualized aluminum deposits according to their presence (a) insideversus outside the brain cells and (b) in the two types of brain tissue (grey matterversus white matter).
For this component, fixed tissue samples were available for the same five donors plus an additional five donors diagnosed with ASD, including two females (ages 13 and 29) and three males (ages 14, 22 and 29).
What the research team found was startling. The study’s quantitative arm documented “consistently high” aluminum levels representing “some of the highest values for brain aluminum content ever measured in healthy or diseased tissues.” Specifically:
All five individuals had at least one brain tissue with a “pathologically significant” level of aluminum, defined as greater than or equal to 3.00 micrograms per gram of dry brain weight (μg/g dry wt). (Dr. Exley and colleagues developed categories to classify aluminum-related pathology after conducting other brain studies, wherein older adults who died healthy had less than 1 μg/g dry wt of brain aluminum.)
Roughly two-thirds (67%) of all the tissue samples displayed a pathologically significant aluminum content.
Aluminum levels were particularly high in the male brains, including in a 15-year-old boy with ASD who had the study’s single highest brain aluminum measurement (22.11 μg/g dry wt)—many times higher than the pathologically significant threshold and far greater than levels that might be considered as acceptable even for an aged adult.
Some of the elevated aluminum levels rivaled the very high levels historically reported in victims of dialysis encephalopathy syndrome (a serious iatrogenic disorder resulting from aluminum-containing dialysis solutions).
The study’s qualitative findings were equally concerning:
Across the 10 donors, the investigators identified 150 aluminum deposits. All 10 donors had aluminum deposits in at least one tissue.
Aluminum deposits were markedly more prevalent in males than females (129 deposits in seven males, averaging over 18 deposits each, versus 21 deposits in three females, for an average of 7).
In males, most aluminum deposits were inside cells (80/129), whereas aluminum deposits in females were primarily extracellular (15/21). The majority of intracellular aluminum was inside non-neuronal cells (microglia and astrocytes).
Aluminum was present in both grey matter (88 deposits) and white matter (62 deposits). (The brain’s grey matter serves to process information, while the white matter provides connectivity.)
The researchers also identified aluminum-loaded lymphocytes in the meninges (the layers of protective tissue that surround the brain and spinal cord) and in similar inflammatory cells in the vasculature, furnishing evidence of aluminum’s entry into the brain “via immune cells circulating in the blood and lymph” and perhaps explaining how youth with ASD came to acquire such shockingly high levels of brain aluminum.
The Importance of Glial Cells
There are three broad categories of non-neuronal (glial) cells, including astrocytes (which support neuronal signaling), oligodendrocytes (which create myelin) and microglia (responsible for repairing damage). In discussing their results, Dr. Exley’s team comments that the intracellular location of most of the aluminum in these non-neuronal cells was the “standout observation” for ASD.
…environmental factors can alter microglia function, negatively affecting brain development and synaptic connectivity; when this occurs during important developmental periods, there may be ‘consequences throughout life.’
Unlike other brain cells, the microglia (which represent about 10% of brain cells) are dedicated immune cells. Microglia also play a key role in the process known as synaptic pruning that takes place during vital phases of cognitive development in early childhood as well as adolescence, continuing into the late 20s.
This process, which some observers have likened to “neural spring cleaning,” allows the maturing brain to shed “weak or redundant [neuronal] connections.” Given this and other important microglial functions, the microglia have attracted considerable research attention as key players in brain disease, including autism.
(Astrocytes also have implications for autism, given the role of astrocyte dysfunction in seizures — a condition that is frequently comorbid with ASD.)
A pivotal review article published in 2017 observes that “microglia are now known to be active participants in brain function and dysfunction” and notes that “aberrant [synaptic] pruning during critical developmental periods could contribute to neurodevelopmental disorders.”
Evidence suggesting that the microglia are dysfunctional in ASD includes findings from postmortem ASD brain studies showing “altered microglial counts, morphology, and neuronal interaction” as well as altered expression of microglia-specific genes.
It is clear to many researchers that environmental factors can alter microglia function, negatively affecting brain development and synaptic connectivity; when this occurs during important developmental periods, there may be “consequences throughout life.”
Aluminum exposure undoubtedly constitutes a dangerous environmental exposure, and Dr. Exley observes that “microglia heavily loaded with aluminum…will inevitably be compromised.”
The Most Pervasive Exposure to Aluminum
The study’s results strongly suggest that aluminum is entering the brain in ASD via cells that have become loaded up with aluminum in the periphery. Where is the aluminum coming from? One of the most pervasive routes of modern-day exposure to neurotoxic aluminum is via aluminum adjuvants in vaccines.
(Vaccine manufacturers use aluminum adjuvants to intensify the vaccine recipient’s immune response.)
Elsewhere, Dr. Exley has described the “migratory capabilities” of aluminum-based adjuvants “at sites distant to the injection site,” including the brain.
The extreme levels of aluminum found in the brains of the study’s teenage donors have alarming implications for the entire generation of highly aluminum-vaccinated children.
In the ASD brain paper, Dr. Exley and coauthors point out that the “burgeoning” use of aluminum-adjuvant-containing childhood vaccines “has been directly correlated with increasing prevalence of ASD.”
A 2011 study by Lucija Tomljenovic and Christopher Shaw confirms that aluminum-containing vaccines are having crippling neurological consequences.
Their analysis shows that children from countries where ASD prevalence is highest have the highest exposure to aluminum from vaccines; moreover, children’s increased exposure to aluminum adjuvants over the two decades starting in the 1990s significantly correlates with the increase in ASD prevalence in the U.S. Counting the shots now pushed during pregnancy, highly vaccinated American children may receive up to 73 total vaccine doses by age 18, including multiple rounds of injected aluminum.
Crucially, Dr. Exley and coauthors note that what “discriminates [their] data from other analyses of brain aluminum in other diseases is the age of the ASD donors” [emphasis added].
The extreme levels of aluminum found in the brains of the study’s teenage donors have alarming implications for the entire generation of highly aluminum-vaccinated children.
Given that it is no longer unheard of to see Alzheimer’s being diagnosed in people who are in their 20s, 30s, or 40s, it is not unreasonable to worry that a catastrophic new wave of AD may be about to compound children’s already heavy burden of ASD and other neurological disorders.
Recognizing the risks, numerous researchers have called for a halt to the use of aluminum salts in vaccines. The powerful results of this study underscore the urgency of heeding this plea as well as eliminating exposure to other sources of neurotoxic aluminum.
“Our citizens should know the urgent facts…but they don’t because our media serves imperial, not popular interests. They lie, deceive, connive and suppress what everyone needs to know, substituting managed news misinformation and rubbish for hard truths…”—Oliver Stone
Internal Site Commenting is limited to members.
Disqus commenting is available to everyone.