Patient with Parkinson's disease working on hand coordination

Parkinson's Disease

Parkinson's Disease

Last Section Update: 11/2023

Contributor(s): Debra Gordon, MS; Shayna Sandhaus, PhD; Stephen Tapanes, PhD

1 Overview

Summary and Quick Facts for Parkinson's Disease

  • Parkinson’s disease is a neurological disorder that causes movement problems and can progress into dementia. It’s caused by the loss of cells in the brain that produce dopamine, a neurotransmitter that controls movement and coordination. Several factors can contribute to the loss of dopamine-producing cells.
  • This protocol will help you understand the causes of Parkinson’s disease and what treatments are available. Learn about some emerging treatment strategies and how some supplements and dietary choices may help protect dopamine neurons.
  • Supplementation with coenzyme Q10 (CoQ10) and creatine has been shown to delay cognitive decline in people with Parkinson’s disease.

What is Parkinson's Disease?

Parkinson’s disease is a degenerative disease of the central nervous system resulting from depletion of dopamine-producing cells in the brain. While the underlying cause of the disease is not clearly understood, depletion of dopamine-producing cells may be exacerbated by oxidative stress, inflammation, and mitochondrial dysfunction. Parkinson’s patients generally experience decline in motor function and eventually cognitive decline and dementia.

Parkinson’s may manifest as a primary condition or secondary to another condition, such as a brain tumor, exposure to toxins, or after a viral infection.

Existing conventional treatments do not slow or reverse the course of the disease. However, natural interventions such as coenzyme Q10 and creatine may support neuronal health and promote mitochondrial function.

What are the Risk Factors for Parkinson’s Disease?

  • Family history/genetic predisposition
  • Exposure to pesticides or other toxins like carbon monoxide
  • Brain tumor
  • Viral encephalitis
  • AIDS
  • Chronic constipation
  • Repeated blows to the head (eg, professional fighters or football players)
  • Certain medications
  • Stroke

What are the Signs and Symptoms of Parkinson’s Disease?

Note: Symptoms of Parkinson’s generally progress slowly. Tremors are often the first sign.

  • Tremors, often in the hand
  • Muscle cramping or rigidity
  • Pain throughout the body
  • Slow movements
  • Incontinence or constipation
  • Difficulty swallowing and/or controlling saliva
  • Dizziness
  • Sleepiness
  • Depression and/or anxiety
  • Hallucinations or frightening dreams
  • Cognitive decline and/or dementia

What are Conventional Medical Treatments for Parkinson’s Disease?

  • Levodopa, or L-DOPA
  • Monoamine oxidase-B inhibitors (eg, selegiline and rasagiline)
  • Catechol-O-methyltransferase inhibitors
  • Dopamine agonists
  • In extreme cases, ablative surgery or deep brain stimulation

What are Emerging Therapies for Parkinson’s Disease?

  • Amantadine, an antiviral drug, may help reduce the side effects of L-DOPA
  • Nicotine
  • Granulocyte colony-stimulating factor (G-CSF), a growth factor that promotes creation of new neurons, has shown promising results in animal models.
  • Stem cell replacement therapy
  • Cognitive-behavioral therapy

What Dietary and Lifestyle Changes Can Be Beneficial for Parkinson’s Disease?

  • Physical therapy and exercise
  • For patients on L-DOPA, protein meal distribution may be recommended (ie, eating dietary protein separate from dosing with L-DOPA)

What Natural Interventions May Be Beneficial for Parkinson’s Disease?

  • Coenzyme Q10 (CoQ10). Patients with Parkinson’s appear to be deficient in CoQ10. Supplementation may slow the progressive deterioration of function in Parkinson's disease (Shults 2002) and have a neuroprotective effect.
  • Creatine. Creatine deficiency is associated with neurological damage. Some studies indicate supplementation may slow disease progression.
  • Omega-3 fatty acids. Levels of omega-3 fatty acids in nerve cell membranes decrease with age, oxidative stress, and in neurodegenerative disorders such as Parkinson's disease. Supplementation may favorably modify brain function and protect brain health.
  • Coffee. Coffee consumption is linked to a reduced risk of developing Parkinson’s disease. Coffee extracts have been shown to act via mechanisms similar to some pharmaceutical Parkinson's therapies.
  • Nicotinamide riboside. Decline in NAD+, a cofactor critical for regulating cellular energy balance, is associated with Parkinson’s disease. Administering nicotinamide riboside, an NAD+ precursor, may offer benefits in Parkinson’s patients.
  • B vitamins. B vitamins (eg, folate, B12, B6, etc.) lower homocysteine levels. Many studies have shown B vitamins to have beneficial effects in Parkinson’s patients, and supplementation may be recommended in patients taking L-DOPA.
  • Vitamin D. Parkinson’s patients tend to have lower serum vitamin D levels than those without the disease. Several studies have shown that higher levels of vitamin D protect against the onset of Parkinson's disease symptoms.
  • Other natural interventions that may benefit Parkinson’s patients include carnitine, green tea, resveratrol, wild green oat extract, pyrroloquinoline quinone (PQQ), and others.

2 Introduction

Parkinson's disease is a degenerative disease of the central nervous system resulting from depletion of dopamine-producing cells in a region of the brain called the substantia nigra. A variety of genetic and environmental factors underlie this loss of brain cells. However, emergent research implicates oxidative stress, inflammation, and dysfunctional mitochondria as major contributors to neurodegeneration in Parkinson's disease.

Up to one million Americans live with Parkinson's disease, with 60,000 new cases being diagnosed each year. Men are more likely to be affected than women, and the risk increases substantially after age 50–60; however, one in 20 patients is diagnosed under the age of 40.1,2

Progression of the disease usually leads to characteristic symptoms such as tremors, muscle rigidity, bradykinesia (slowness and difficulty with movements), poor balance, sleep disturbances, and loss of coordination; eventually, cognitive decline occurs, and, in advanced disease, dementia arises.

Conventional medical approaches to treating Parkinson's disease aim to replace the lost dopamine, but fall short of addressing the ongoing destruction of dopaminergic neurons. Over time, the ability of medications to replenish dopamine levels becomes overwhelmed by further loss of dopaminergic cells. Moreover, the pharmaceutical drugs typically used to alleviate symptoms of Parkinson's disease are laden with debilitating side effects and often worsen affection over time. Thus, the prognosis for Parkinson's disease patients relying on conventional treatment remains limited.

The mainstream medical establishment has failed to recognize the urgent need to address the multiple, interrelated pathological features of Parkinson's disease in order to prevent further neuronal loss and slow disease progression.

Scientific innovation has led to the realization that natural compounds and some underappreciated pharmaceutical compounds can synergize to support mitochondrial function, suppress inflammation, ease oxidative stress and may improve outlook for Parkinson's disease patients.

Life Extension's approach encompasses a regimen combining conventional therapeutics to ease symptoms and innovative natural ingredients along with state-of-the-art pharmaceuticals to reduce the destruction of dopaminergic neurons. This approach offers Parkinson's disease patients a chance for symptomatic improvement and enhanced quality of life.

3 Brief History, Classifications, and Risk Factors

Dr. James Parkinson first described the motor system disorder known today as Parkinson's disease in an 1817 paper entitled "An Essay on the Shaking Palsy."3 In his report, Dr. Parkinson described several characteristic traits, including an abnormal posture and gait, and partial paralysis with muscle weakness; he also described the progression of the disease. The contribution of more clearly defining the condition, theretofore known as paralysis agitans, led to the adoption of Dr. Parkinson's last name as the moniker that remains with us today.

Since 1817, medical advancements have helped us establish a much greater understanding of Parkinson's disease. Today, clustered symptoms like tremor at rest, stiffness, slowed movement, and postural instability are classified, based upon their cause, into different categories.

Parkinson's Disease (Primary Parkinson's)

This is the most common form of the disease; what most of us think of when we hear the term "Parkinson's." Primary Parkinson's disease has no clear external cause, and is therefore classified as idiopathic or without cause (arising spontaneously). Recently, however, several genes directly tied with the development of Parkinson's disease have been identified. This has led to the classification of heritable Parkinson's disease of genetic origin as familial Parkinson's disease, while Parkinson's disease that arises independently of genetic predisposition is referred to as sporadic Parkinson's disease.

Despite the fact that conventional medical dogma holds tightly to the notion that primary Parkinson's disease truly lacks an identifiable cause (other than genetics in familial Parkinson's disease), metabolic phenomena, such as oxidative stress, mitochondrial fatigue, and other age-related abnormalities are linked with the death of dopamine-producing neurons.4

Exposure to pesticides may substantially increase risk for Parkinson's disease.5-10 In one study, higher pesticide exposure increased Parkinson's disease risk three-fold.10 Numerous epidemiological studies have confirmed the association.6,11 Toxin-induced Parkinson's symptoms may be classified as secondary, rather than primary Parkinson's.4,12

Interestingly, pesticides seem to accumulate in the dopaminergic tract, where they inhibit mitochondrial function and lead to neuronal death.7,13 Dopaminergic neurons are particularly susceptible to the pesticide dieldrin, which is no longer in use in the United States, but remains ubiquitous due to environmental contamination.14 In addition to acting as neuronal and mitochondrial toxins, some pesticides also impair the breakdown of protein aggregates, like Lewy bodies.15

Several lines of evidence suggest that a genetic inability to properly detoxify environmental toxicants may predispose some individuals to Parkinson's disease.16,17

In addition, those who experience constipation throughout their lives appear to be at increased risk.18 In one study, constipation documented in medical records as much as 20 years before disease onset was associated with a significantly increased risk.19 Some researchers believe that this may be related to intake of drinking water—lower water intake appears to be a risk factor as well.20 This may be linked to reduced elimination of water-soluble toxins.

Due to the strong association between pesticides, and other environmental toxins, with Parkinson's disease, readers are strongly encouraged to review Life Extension's “Metabolic Detoxification” protocol.

Parkinsonian Syndrome (Secondary Parkinson's)

Other forms of Parkinsonism can occur as a secondary effect of brain tumor, drugs, toxins (eg, carbon monoxide poisoning), post encephalitis (viral infectious disease, "sleeping sickness"). For example, another cause of Parkinsonism is brain damage sustained by repeated blows to the head such as suffered by professional prize fighters and athletes in high-impact sports like football. Traumatic events, infections, use of certain medications, etc. can all damage the dopaminergic cells within the midbrain and lead to the same symptoms as primary Parkinson's disease.

For example, the defining basis for Parkinsonism due to encephalitis (brain inflammation) was a worldwide influenza pandemic in 1917. After recovering from this illness, many patients developed Parkinson's disease years later.21 Acquired immunodeficiency syndrome (AIDS) may also lead to Parkinsonism.22 Resuscitation from cardiac arrest (due to temporary lack of oxygen supply to the brain), and stroke can lead to Parkinsonism as well.23

Several centrally acting drugs, especially those that exert an effect on the dopamine system within the brain, such as antipsychotics, frequently induce secondary Parkinsonism after sustained chronic use. In fact, drug-induced Parkinsonism is a well-documented phenomenon.24-26 Some antidepressants and calcium channel blockers, and the antiarrhythmic drug amiodarone, can lead to Parkinsonian tremors as well.26 Several illicit drugs can cause Parkinsonism as well.

Some diseases or disorders considered to cause Parkinsonian syndromes include multiple system atrophy (MSA), progressive supranuclear palsy (PSP), corticobasal degeneration (CBGD), and Pick's disease.

4 Signs, Symptoms, and Diagnosis

Dopamine is a neurotransmitter that, among other functions, allows messages to be sent to regions of the brain responsible for coordinating movement. When dopamine levels decline, due to the death of dopaminergic cells, these messages no longer reach their destination, and so the regions of the brain that control movement no longer function properly. This results in loss of conscious control of movement, and, in advanced Parkinson's disease, loss of control over several other bodily functions.

The onset and course of Parkinson's disease may be different for each patient. For example, while tremor is evident in most patients, some may not experience movement complications until the disease has advanced considerably.

Initial symptoms of primary Parkinson's disease typically develop slowly and randomly as the supply of dopamine dwindles over time. In some cases, symptoms do not appear until approximately 70% of the dopaminergic cells in the substantia nigra are already destroyed.2

Motor Symptoms

The onset of a slight tremor, usually in the hand, which increases in intensity over time, is often the initial sign of Parkinson's. However, roughly 30% of patients do not develop a tremor. Parkinson's patients often experience muscle rigidity or cramping that can be painful—movements as simple as turning over in bed or buttoning a shirt can become arduous, and as the disease advances, nearly impossible. Progression of Parkinson's disease leads to slowness of movements, which can cause a great deal of frustration for patients who cannot move as quickly as they would like.

"Freezing" is a frequently reported motor symptom in advancing Parkinson's. This involves the sudden onset of the inability to move at all; patients sometimes describe freezing as feeling as if their feet are stuck to the floor. Freezing is temporary and usually lasts from a few seconds to a few minutes.

Non-Motor Symptoms

Dopamine is involved in a number of functions beyond control of movement, so loss of dopaminergic neurons (and other neurons in late-stage Parkinson's) can cause several non-motor symptoms as well. However, non-motor symptoms usually develop at later stages of disease progression; nonetheless, they can be equally as debilitating as motor symptoms for many patients.

Patients with advanced Parkinson's disease may experience a variety of non-motor symptoms. These can include incontinence, constipation, difficulty swallowing, inability to control saliva, dizziness, which can lead to falls, excessive daytime sleepiness, intense frightening dreams, depression and/or anxiety, and hallucinations.2 In addition, Parkinson's disease can cause perceptible pain throughout the body, which is sometimes severe.

Dementia

Dementia and related cognitive decline is a major concern among those with advanced Parkinson's disease; up to 75‒80% of those with Parkinson's develop dementia near the end of their life.27,28 In addition to loss of dopaminergic neurons, cholinergic neurons are also at risk. Cholinergic neurons produce a neurotransmitter called acetylcholine, which is important for cognitive function. The accumulation of protein aggregates (clumps of dysfunctional proteins) known as Lewy bodies within cholinergic neurons is a common characteristic of Parkinson's disease.

As Lewy bodies accumulate inside neurons, the cells can no longer function, and eventually die. Loss of acetylcholine leads to diminished attention span, blunted sensory perceptions, loss of arousal and structural changes in the synaptic junctions (the connections between neurons through which they communicate using chemical and electrical signals). Loss of acetylcholinergic signaling is thought to be associated with memory deficits in Alzheimer's disease as well, though the exact mechanisms are complex.29

Two subsets of dementia exist in the context of Parkinson's disease, Parkinson's disease dementia (PDD) and Dementia with Lewy bodies (DLB). The distinction of the two is quite subjective and largely based upon the time of dementia diagnosis in relation to onset of motor symptoms. Whether or not the two dementias are truly separate entities, or simply manifestations of different points along the "Lewy body spectrum," is a hotly debated topic.30

Diagnosis

Clinicians must rely on clinical experience, interpretation of symptoms, and evaluation of medical history in order to tentatively diagnose a patient as having Parkinson's disease. This is because there are no lab tests available that definitively diagnose Parkinson's disease. Parkinson's disease is a diagnosis of exclusion; in other words, the physician will first rule out other possible diagnoses before assuming Parkinson's.

If Parkinson's is suspected because the patient is exhibiting signs such as a tremor on one side of their body, or rigidity with loss of postural reflexes, oftentimes L-DOPA, a drug used to treat Parkinson's symptoms, is administered. If L-DOPA causes the symptoms to subside, the diagnosis of Parkinson's disease can be made more confidently, yet still not definitively.

Due to the elusive nature of a definitive Parkinson's disease diagnosis, patients should be reevaluated regularly to make sure that their symptoms are not due to another neurological disorder that causes similar symptoms.

5 Causes, Pathological Mechanisms, and Lessons from Biology

Genetics—Familial Parkinson's

Roughly 15% of Parkinson's disease patients have a first-degree relative who also has/had Parkinson's disease; this suggests that genetics play a consequential role in the development of familial Parkinson's disease.31 Roughly nine genetic mutations have been associated with Parkinson's disease; of these, six have been particularly well characterized.31,32 Mutations in these genes are generally associated with early onset Parkinson's disease, which is diagnosed before age 40; Parkinson's disease of genetic origin is sometimes diagnosed in childhood.

Mutations in the following genes are associated with an increased risk of Parkinson's disease:

  • SNCA33-36
  • LRRK237
  • PARK238
  • PINK139-42
  • PARK743-47
  • ATP13A248-51

Additional research is required to fully elucidate the role of genetics in Parkinson's etiology; it is likely that several additional genes involved in the pathology will be identified in the coming years. Treatments based upon genetic therapy are likely to become more widespread and therapeutic as scientific knowledge progresses.

Genetic Testing

Genetic testing for mutations known to be associated with Parkinson's disease is available through genetics health care professionals. Specifically, tests are available that check for mutations in PINK1, PARK7, SNCA, and LRRK2. Although the testing is expensive, and accuracy is a potential concern, those individuals with a family history of Parkinson's disease are encouraged to discuss genetic testing with their healthcare provider.

The National Human Genome Research Institute, a division of the National Institutes of Health, has compiled further information about the role of genetics and genetic testing in Parkinson's disease. This resource can also assist with the location of a genetic counselor near you. Their website is: http://www.genome.gov/10001217#4.

Individuals found to have a mutation in one or more of the genes linked to Parkinson's, as well as those with a family history of Parkinson's, should consult a Parkinson's disease specialist, and initiate nutritional and lifestyle strategies to combat neurodegeneration.

Mitochondrial Dysfunction

A flurry of emergent research has linked mitochondrial dysfunction to the pathogenesis of Parkinson's disease. Mitochondrial dysfunction results in impaired ATP generation, loss of cellular repair mechanisms, and cellular inefficiency.

As mitochondria become dysfunctional they generate large quantities of free radicals, which contribute to oxidative stress that, in turn, causes further mitochondrial dysfunction. Concurrently, loss of mitochondria to oxidative damage means fewer mitochondria are available to meet the energy demands of the cell to repair damaged components. The cascade of mitochondrial dysfunction, oxidative stress, and loss of mitochondria form a continuity that ultimately leads to cell death.52,53

Numerous studies have clearly identified mitochondrial dysfunction as a central pathological feature of both genetic and sporadic Parkinson's disease.54,55 Moreover, many of the genes that confer predisposition to familial Parkinson's are intimately related to mitochondrial function; much of the neuronal death in Parkinson's of genetic origin is due to mitochondrial dysfunction, and impaired mitophagy.56-58 While several factors, including exposure to environmental toxins,56,59,60 also contribute to mitochondrial dysfunction in the substantia nigra, age-related mutations in mitochondrial DNA are thought to be a primary culprit.60,61 Alarmingly, dopamine itself, and L-DOPA, may contribute to mitochondrial toxicity in dopaminergic neurons.62-64

Mitophagy, Lewy Bodies, and alpha-Synuclein

Damaged mitochondria are continually being cleared from within the cell through a process calledmitophagy. Mitophagy, a type of autophagy, is a kind of cellular recycling system that clears damaged mitochondria before they can accumulate and cause cellular dysfunction. However, age-related mutations in mitochondrial DNA, which cause mitophagy to become less efficient, coupled with an ever-intensifying propensity for endogenous and environmentally mediated mitochondrial damage cause the neuronal mitophagic system to become overwhelmed.57,65 Over time, damaged mitochondria build up inside the neuron, leading to cell death. Not surprisingly, several of the genetic mutations linked to familial Parkinson's disease cause disturbances in mitophagy.57,58

Another toxic byproduct of mitochondrial dysfunction and impaired mitophagy is the formation of Lewy bodies. Lewy bodies form as reactive oxygen species derived from dysfunctional mitochondria damage structural components of the cell called microtubules. As microtubules are damaged, they release a protein called alpha-synuclein. The loose alpha-synuclein proteins then group together, or aggregate, and form a toxic mass (a Lewy body) that further damages the cell. Moreover, alpha-synuclein has been shown to directly interfere with mitochondrial function and inhibit ATP synthesis, furthering the spread of mitochondrial dysfunction in the brains of Parkinson's disease patients.66-68 Over time, Lewy bodies spread to neighboring cells, damaging neurons within the vicinity of a dead or dying neuron.69

Lewy bodies share some characteristics with toxic proteins that develop in the brains of patients with Alzheimer's disease and other neurodegenerative diseases, primarily in that they cannot be broken down and cleared from the cell by normal autophagic (cellular house cleaning) actions.

The Role of Inflammation in Parkinson's Disease

Inflammatory responses contribute to the perpetuation of neurodegeneration in Parkinson's disease. The brain contains immune cells called microglia, which are known to be activated in Parkinson's disease.70,71 Upon activation, microglia release inflammatory cytokines that can spread to nearby healthy neurons and cause degeneration. Dopaminergic neurons in the substantia nigra, the brain region most affected by Parkinson's disease, express receptors for an inflammatory cytokine called tumor necrosis factor-alpha (TNF-α), which suggests that excess TNF-α released by nearby activated microglia may damage nigral dopaminergic cells.

Elevated cytokines in the brain of those with Parkinson's disease is a consequence of neurodegeneration.72 In experimental models, exposure to the neurotoxin MPTP (a chemical used to induce Parkinson's disease in experiments) leads to death of dopaminergic neurons. Interestingly, in monkeys, inflammation is increased even years after initial exposure to MPTP.71 This suggests inflammation, once initiated, has long-term consequences in Parkinson's disease.

As dopaminergic cells succumb to either environmentally or genetically induced mitochondrial dysfunction, they release free radicals. These free radicals then activate nearby microglial cells, which in turn, excrete inflammatory cytokines that bind to and damage nearby dopaminergic neurons. This positive feedback loop may continue over years or even decades and slowly contribute to the loss of dopaminergic neurons that leads to Parkinson symptoms.72,73

Epidemiological studies on the use of anti-inflammatory drugs and the risk of Parkinson's onset are conflicting. Some studies suggest a protective role of ibuprofen, but not other anti-inflammatory drugs.74 However, a large study published in the British Medical Journal involving over 22,000 subjects found no association between use of any NSAID reduced risk.75 These findings reinforce the notion that, rather than initiating dopaminergic cell death, inflammation may perpetuate it, thus contributing to Parkinson's disease progression. Life Extension believes that suppressing inflammation may slow disease progression in Parkinson's disease patients.

6 Conventional Medical Treatment

For decades, the conventional standard of care for Parkinson's disease has focused on symptomatic relief. Pharmaceutical treatments for Parkinson's accomplish this by either increasing dopamine levels or mimicking its action. While conventional therapeutics are indispensable for improving quality of life in Parkinson's patients, they do not provide fundamental neuroprotection or support for neuronal mitochondria. Thus, mainstream pharmaceutical treatments cannot be expected to address the underlying cause of disease progression—neurodegeneration.

Treatment with L-DOPA causes patients to be less responsive to the medication over time and can evoke a number of adverse side effects. However, careful dosing strategies, and utilization of ancillary medications may help limit side effects and maintain the effectiveness of conventional pharmaceutical therapies.

Pharmaceutical treatment of Parkinson's disease symptoms is usually initiated when the patient has already developed some disability for which he/she needs to be treated. This is typically referred to as the initial stage of therapy. The primary goal of treatment during the initial stage is to limit symptoms arising from progression of the disease. However, with time, adverse side effects of the medications arise, which leads into the secondary treatment stage. The aim of the secondary treatment stage is to reduce Parkinson's symptoms, as well as counterbalance the adverse side effects of levodopa.

Levodopa (L-DOPA)/Carbidopa

Since its FDA approval in 1970, Levodopa (L-DOPA) has been a staple for the management of Parkinson's disease symptoms.

L-DOPA (the precursor to dopamine) is metabolized into dopamine in the body by an enzyme called aromatic L-amino acid decarboxylase (AADC). Dopamine itself cannot pass through the protective blood-brain barrier, but L-DOPA can. When L-DOPA is administered orally, a small percentage passes into the brain and is converted into dopamine. This temporary increase in dopamine levels within the brain offers relief of Parkinson's disease symptoms for a short period.

However, the body presents many obstacles that limit the efficiency of oral L-DOPA therapy. First, AADC exists outside the brain as well, which means that the majority of orally administered L-DOPA will be converted into dopamine peripherally (not in the central nervous system). Therefore, L-DOPA is typically administered with an inhibitor of peripheral AADC, called carbidopa. Carbidopa (or another AADC inhibitor) helps preserve orally administered L-DOPA for conversion to dopamine in the brain.

Regrettably, the use of orally administered L-DOPA over time results in diminished production of endogenous (naturally occurring within the body) L-DOPA. L-DOPA therapy is further complicated by the development of movement disorders called dyskinesias after 5–10 years of use in most cases.

Dyskinesias are movement disorders in which neurological discoordination results in uncontrollable, involuntary movements. This discoordination can also affect the autonomic nervous system, resulting in, for example, respiratory irregularities.82 Dyskinesia is the result of L-DOPA-induced synaptic dysfunction and inappropriate signaling between areas of the brain that normally coordinate movement, namely the motor cortex and the striatum.83

With long-term L-DOPA use (usually after about five years), responsiveness declines and dose adjustment is often necessary. This phenomenon leads to fluctuations in the effectiveness of L-DOPA therapy that cause the patient to experience dyskinesia as the post-dose concentration of L-DOPA peaks, and rapid reversion to severe Parkinsonism towards the end of the dosing period.

Several strategies exist for enhancing L-DOPA effectiveness. Some of these include varying combinations of L-DOPA and other medications discussed in this section as well as altering dose timing and amount. Other strategies can involve "rest periods" or "drug holidays" during which the patient abstains from L-DOPA for a short time; as little as skipping a single dose each day may help lessen the damage caused by oxidation products of L-DOPA metabolism and maintain dopamine receptor sensitivity. A patient should never adjust their L-DOPA dose without close supervision by their physician.

Other strategies for stabilizing dopamine levels include combining L-DOPA with inhibitors of enzymes that breakdown dopamine. Medications of this type include monoamine oxidase-B (MAO-B) inhibitors, and catechol-O-methyltransferase (COMT) inhibitors. By combining L-DOPA with COMT and/or MAO-B inhibitors, a physician may be able to reduce the dose of L-DOPA required to relieve symptoms, and widen dose intervals, which is more convenient for the patient.

There are a variety of ways that pharmaceuticals can be combined to deliver optimal effects in each Parkinson's case, but the needs of each patient may vary widely. Therefore, patients should always consult an experienced physician to discuss medication combinations that may be ideal for their unique situation.

L-DOPA can produce several adverse side effects, including:

  • Arrhythmia
  • Gastrointestinal discomfort (taking L-DOPA with low protein snacks may help avoid stomach upset)
  • Breathing disturbances
  • Hair loss
  • Confusion
  • Extreme emotional variability with prevalent anxiety
  • Vivid dreams
  • Hallucinations
  • Impaired social behavior
  • Sleepiness
  • Excessive libido
  • Compulsive behavior (ie, reckless gambling)

L-DOPA-induced elevations in homocysteine, a potentially harmful amino-acid derivative, are another major concern for Parkinson's patients. High levels of homocysteine have been implicated in various cardiovascular diseases, including cerebral small vessel disease, as well as brain atrophy.84,85 A comprehensive review of 16 studies found that elevated homocysteine was associated with dementia and markers of neurodegeneration in patients with Parkinson's patients.86

Parkinson's disease patients taking L-DOPA should read Life Extension's “Homocysteine Reduction” protocol and strive to maintain homocysteine levels of less than 7–8 µmol/L.

L-DOPA Drug Holidays

Regular, chronic use of L-DOPA causes dopamine receptors within the brain to become less sensitive, leading to the eventual need for increased dosages of L-DOPA. Research suggests taking a "drug holiday" from L-DOPA may re-sensitize dopaminergic receptors and lower the patient’s L-DOPA requirements, or at least prevent the need for increasing L-DOPA in the near future. In a three-year study, 15 Parkinson's patients were submitted to a seven-day L-DOPA drug holiday. Within the first six-months following the drug holiday, symptoms improved dramatically, and all of the study subjects were able to maintain a L-DOPA dose regimen of 50–70% of their pre-holiday dose for the entire three-year period.87

Despite these promising results, there are serious risks associated with stopping L-DOPA therapy, one of which is neuroleptic malignant syndrome, a potentially life-threatening situation. Therefore, a drug holiday should only be initiated under the close supervision of a physician. However, at least one study suggests use of amantadine, another drug used to alleviate Parkinson's symptoms, during an L-DOPA drug holiday may limit the severity of side effects associated with stopping L-DOPA therapy. In this study, 12 Parkinson's patients were submitted to a three-day L-DOPA drug holiday, and during that time they were given I.V. infusions of amantadine. The subjects were then started back on the pre-holiday L-DOPA dose and symptomatic improvements lasting up to four months were noted.88

Dopamine Agonists

Another method used to restore dopaminergic signaling in Parkinson's disease is medicating with a dopamine agonist. A dopamine agonist is a drug containing a molecule that binds to and activates dopamine receptors, similar to dopamine itself, thus compensating for low dopamine levels. Dopamine agonists are often used in younger patients, or in very early Parkinson's disease.

Research comparing the results of initial therapy with a dopamine agonists or L-DOPA is conflicting. Some studies suggest that initiating therapy with a dopamine agonist may delay the onset of dyskinesias as the disease progresses, while some seem to indicate that this may not be the case. Other studies suggest initial dopamine agonist therapy delivers results similar to those seen in L-DOPA plus COMT inhibitor therapy.89 Results from a 14-year follow up study found that initial therapy with a dopamine agonist offered no greater benefit over standard L-DOPA therapy in the long term.90

Dopamine agonists pose a greater risk of serious side effects than L-DOPA and are therefore not as tolerable for some patients. Some side effects of dopamine agonists include:

  • Euphoria
  • Hallucinations
  • Psychosis
  • Orthostatic hypotension (low blood pressure upon standing)
  • Increased orgasmic intensity
  • Weight loss
  • Nausea
  • Insomnia
  • Unusual tiredness or weakness
  • Dizziness or fainting
  • Twitching, twisting, or other unusual body movements
  • Pathological addiction and compulsive behavior (ie, hyper-sexuality, gambling)

Selegiline and Rasagiline

Selegiline is a MAO-B inhibitor that, due to its unique chemical structure, also exerts other neuropharmacological actions via its metabolites. By blocking the breakdown of dopamine, selegiline helps compensate for the diminished production of dopamine in Parkinson's disease. This can lead to symptomatic improvement, especially in early-stage Parkinson's.

Numerous clinical trials have confirmed the efficacy of selegiline alone and in combination with L-DOPA in early Parkinson's disease.91-93 One study showed that selegiline was highly effective if initiated within five years of Parkinson's disease diagnosis, but less effective if initiated 10 years or more after diagnosis.91

Selegiline exerts a number of other benefits as well, including maintenance of whole-brain blood flow in depressed Parkinson's disease patients.94 Moreover, selegiline may reduce the formation and toxicity of alpha-synuclein aggregates.95

Rasagiline is a newer generation medication based upon selegiline. Laboratory studies suggest that, in addition to functioning very similarly to selegiline, rasagiline may exert a greater neuroprotective effect.96

Rasagiline was superior to placebo in slowing progression of Parkinson's disease in a cohort of 1,176 early-stage patients. In this study, subjects receiving rasagiline were less likely than those taking placebo to need additional anti-Parkinson drugs to manage symptoms.97 More trials need to be conducted to determine if rasagiline is significantly more effective than selegiline for treating Parkinson's disease.

Selegiline is available via prescription in a clinically studied transdermal patch called Emsam. Selegiline and rasagiline may cause dizziness, dry mouth, sleeplessness, and an overall stimulating effect.

7 Alternative and Emerging Therapies

In addition to the conventional standard of care, which relies heavily on L-DOPA therapy, physicians may sometimes implement other pharmaceutical agents that complement the effects of L-DOPA therapy, or limit its side effects.

Amantadine

Amantadine is an antiviral drug that exerts a number of actions in the brain. Amantadine has been shown in some studies to benefit Parkinson's patients, primarily by reducing the side effects of L-DOPA, or as an adjuvant during L-DOPA drug holidays as mentioned above, though the mechanisms are largely unclear.

In clinical studies, amantadine has been shown to temporarily reduce L-DOPA induced dyskinesia; an effect which dissipates after about eight months.98,99 However, in some patients, discontinuation of amantadine appears to cause a rebound worsening of dyskinesias to an even higher intensity than before its introduction.99

As mentioned earlier in this protocol, at least one study suggests amantadine may suppress side effects of L-DOPA abstinence during a drug holiday.88

Amantadine may ease Parkinson's symptoms in some patients, but should only be initiated under physician supervision.

Nicotine

Within the brain, there exists a grand diversity of neurotransmitter interaction and overlap. One such relationship is that existing between the dopaminergic and cholinergic systems. For example, acetylcholine modulates dopaminergic signaling in the striatum, an area considerably impacted in Parkinson's disease.

Nicotine interacts with the cholinergic system by binding to sites known as nicotinic acetylcholinergic receptors (nAChRs), which influence several functions relevant in Parkinson's disease, including dopamine signaling.100 Moreover, loss of nAChRs accompanies many neurodegenerative diseases, including Parkinson's disease, suggesting that declining cholinergic signaling may be a key etiological feature.101 Several studies indicate that nicotine exerts neuroprotective effects via activation of nAChRs.102 Some data indicates that among the neuroprotective effects of nicotine is the ability to reduce alpha-synuclein aggregation, which may suppress the formation of Lewy bodies.103

Epidemiological evidence suggests that smoking tobacco is associated with a reduced risk of developing Parkinson's disease.104,105 Moreover, transdermal nicotine patches have been shown to improve cognitive functioning in patients with Parkinson's disease.106 Other evidence suggests a therapeutic effect of nicotine in reducing L-DOPA-induced dyskinesias.107

However, the ability of nicotine to improve symptoms and slow progression of Parkinson’s disease is inconclusive. A double-blind, randomized, controlled trial randomly assigned 163 therapy-naïve patients with early Parkinson’s disease, diagnosed within 18 months and in Hoehn and Yahr stage ≤2, to receive transdermal nicotine (7–28 mg/day, titrated to the maximum tolerated dose) or placebo for 52 weeks followed by an 8-week washout period. Patients did not require dopaminergic therapy at the initiation of the trial. The primary endpoint was change in Unified Parkinson’s Disease Rating Scale (UPDRS) at 60 weeks; the secondary endpoint was change in UPDRS at 52 weeks. The primary outcome worsened by 3.5 points in the placebo group and 6.0 points in the nicotine group. The secondary endpoint showed similar results, with a worsening of 5.4 in the placebo group and 9.1 in the nicotine group.325 A separate study examining 40 patients with Parkinson’s disease receiving 90 mg/day (titrated over the course of 11 weeks) of transdermal nicotine for 28 weeks found no significant difference in UPDRS motor scores between treated and non-treated groups after six weeks of washout.326 Other studies have reported a worsening or no improvement in Parkinson’s symptoms for patients administered transdermal nicotine compared with placebo.327,328

In summary, while there is experimental evidence supporting nicotine's neuroprotective effect on dopaminergic neurons, clinical trials have yet to provide convincing evidence of its efficacy in slowing Parkinson’s disease progression. The intriguing epidemiological observations and some promising preclinical data suggest further research, with a focus on dosing and delivery systems, may be warranted to fully understand nicotine's potential role in Parkinson’s disease prevention or treatment.

Granulocyte Colony-Stimulating Factor (G-CSF)

G-CSF is a signaling glycoprotein (produced in several tissues) that stimulates the production and differentiation of white blood cells, thereby playing a significant role in immune system function. Recombinant G-CSF is frequently given to chemotherapy patients to restore levels of white blood cells that have been suppressed by treatment.

The interaction of G-CSF with the immune system is very complex. However, current evidence suggests that besides stimulating white blood cell generation, it pushes the immune system towards a less autoreactive, anti-inflammatory TH2 phenotype rich in T-regulatory cells.108 Due to this unique action, G-CSF may be of benefit in diseases in which inflammation contributes to the pathology.

Interestingly, receptors for G-CSF are expressed in neurons throughout the central nervous system and activation of those receptors (by G-CSF) stimulates neurogenesis and protects neurons from damage.108,109

In animal models of both Alzheimer 's disease and Parkinson 's disease, subcutaneous injections of recombinant human G-CSF suppressed inflammation in brain regions centrally involved in the pathology of each disease and stimulated the formation of new synapses.110-112 In these studies, mice treated with G-CSF performed much better on cognitive tests than those not treated with G-CSF. These findings are very exciting and hold promise for future research.

Stem Cells and Cell Replacement Therapy

The hallmark of Parkinson's disease is loss of dopaminergic neurons in the substantia nigra. Therefore, many therapeutic approaches have aimed at replacing lost neurons in this region using cell replacement therapy, or stem cell therapy. These therapies are largely experimental as of the current time and no large-scale clinical trials have been conducted as of yet. In fact, small-scale clinical trials have shown that benefit of replacing dopamine neurons may be questionable, and that the therapy caused severe dyskinesias in some subjects.113

Another major challenge associated with cell replacement therapy is ensuring survival of transplanted neurons. So far, this has proven extremely difficult.114 However, further studies are underway, and advancements in research may allow for widespread use of these therapies in the not-too-distant future.

Focused Ultrasound and Parkinson's Disease

Focused ultrasound (FUS) is an FDA-approved noninvasive, therapeutic technology with the potential to improve quality of life in Parkinson’s Disease patients that experience severe tremors, especially among those for whom medications do not work well. FUS uses ultrasound waves, which carry energy through tissue and can be focused precisely to ablate tissue and cells contributing to tremors.300 FUS is also capable of temporarily disrupting the blood-brain barrier, allowing therapeutics to enter the brain and undesired materials to depart from the brain.301

FUS has the potential to achieve symptomatic relief by causing lesions in the brain to disrupt circuits associated with tremor. Some targets include the thalamus, subthalamic nucleus, globus pallidus, and pallidothalamic tract, which have been implicated in parkinsonian tremor, dyskinesia, and akinesia.302 A 2020 clinical trial randomized 40 participants with Parkinson's disease who had motor signs not fully controlled by medication or who were ineligible for deep-brain stimulation in a 2:1 ratio to receive FUS of subthalamic nuclei or a sham procedure as control. This trial found that FUS in one hemisphere improved motor features of Parkinson’s disease as evidenced by a decrease in Unified Parkinson’s Disease Rating Scale (UPDRS) scores, although some adverse events were reported including speech and gait disturbances and weakness on the treated side.303 The clinical benefits of FUS have been corroborated in multiple clinical trials in which the majority of treated patients reported significant improvements in their tremor, dyskinesia, or akinesia.304

Currently, FUS is limited to being used to treat only one side of the brain, which results in unilateral effects.300 Despite this limitation, FUS is gaining popularity as a therapy since it is a single noninvasive technique that does not require subsequent procedures.

Deep Brain Stimulation and Parkinson's Disease

Deep brain stimulation (DBS) is a surgical procedure in which electrodes are inserted into a target area of the brain to modulate abnormal electrical activity of cells.305 In fact, some consider it to be the most important therapeutic advance in treating Parkinson’s disease symptoms since the development of levodopa. DBS was approved to treat Parkinson’s disease tremor in 1997, then in 2002 for treatment of advanced Parkinson’s disease syndromes, and finally in 2016 for the treatment of early stages of Parkinson’s disease. DBS is capable of providing relief from symptomatic Parkinson’s disease, but it does not slow disease progression.306 Further, its benefits are mostly limited to tremor, dyskinesia, and akinesia; it has not been reliably demonstrated to ameliorate non-motor issues (eg, speech or cognitive problems) that may significantly impact patient’s quality of life.307,308

Various clinical trials have demonstrated the efficacy of DBS in lessening motor symptoms associated with Parkinson’s disease.309 Particularly, DBS as a combination therapy with traditional medications, such as levodopa, may result in an additive effect while also helping lessen side effects associated with dopaminergic medications.310 A five-year, randomized, single-blind trial of patients with early-stage Parkinson’s disease examined the effect of bilateral DBS of the subthalamic nucleus (STN) in combination with levodopa compared with levodopa alone. Patients treated with combination therapy required lower daily dosages of levodopa and had significantly reduced odds of worsening rest tremor compared with levodopa alone.311 A systematic review and meta-analysis of eight randomized controlled trials found DBS led to significant improvements in UPDRS for patients and a reduction in medication dose, although this was accompanied by a significantly higher risk of serious adverse events.309 Further, a meta-analysis of 13 studies found that although the motor benefits achieved via DBS of the globus pallidus internal (GPi) and STN were similar, DBS of the STN allowed for a greater reduction in medication.312 

Potential risks arising from electrode implantation for DBS include surgical complications such as bleeding, stroke, and infection. The benefits of DBS, such as improvements in motor function and dyskinesia, are thought to last for 5‒10 years, although the magnitude of these improvements tends to decline over time.313

Cognitive-Behavioral Therapy

Parkinson's disease is often accompanied by comorbid psychological disturbances such as depression and/or anxiety, and psychosis (a potential side effect of anti-Parkinson medications). Treatment of psychological disturbances is limited, to some degree, due to potential interactions between pharmaceuticals used to treat Parkinson's and those used to treat other psychological conditions.

Cognitive-behavioral therapy offers a highly effective drug free alternative for relieving psychological disturbances in Parkinson's disease patients. In one study, depressed Parkinson's patients were either clinically monitored or engaged in cognitive-behavioral therapy for just over three years. While a mere 8% of patients undergoing clinical monitoring experienced improvements in their depressive symptoms, significant improvement was noted in 56% of those engaged in cognitive-behavioral therapy.116

In addition to the psychological benefits, cognitive-behavioral therapy may be effective for the treatment of some physical symptoms of Parkinson's disease. A 2011 study found that in patients older than 50 years, cognitive-behavioral therapy led to a significant reduction in the incidence of urinary incontinence.117

Several different types of cognitive-behavioral therapy are available and different styles may be appropriate in some cases while inappropriate in others. Patients with Parkinson's disease may benefit from cognitive-behavioral therapy and therefore, should discuss this option with their physicians.

Physical Therapy and Exercise

Parkinson's patients are prone to motor disturbances, such as poor balance and a greater chance of falling, which can lead to decreased mobility. As the disease progresses, engaging in structured physical therapy or exercise may be an effective way of maintaining balance and avoiding falls.118

Moreover, an array of studies has shown that exercise and physical activity in general exert substantial supportive effects upon brain structure and function. In fact, physical activity is associated with a decreased propensity for aging adults to develop dementia, a common problem in Parkinson's disease.119 Experimental Parkinson's disease models demonstrate that physical activity provides neuroprotection and promotes mitochondrial integrity.120

Staying active is very important for Parkinson's disease patients. Those not engaged in regular physical activity are encouraged to speak with their healthcare provider about initiating a structured exercise or physical therapy regimen. A target goal of 75% maximum age adjusted heart rate for a minimum of 20 minutes at least three times per week is ideal. However, this may not be possible for advanced Parkinson's disease patients.

Simvastatin and Parkinson's Disease

Some research suggests the cholesterol lowering drug simvastatin (Zocor) may provide neuroprotection in Parkinson's disease, although the literature overall is inconsistent and inconclusive. Statin drugs do more than just lower cholesterol—they are also anti-inflammatory agents. In fact, many researchers suggest some of the cardiovascular benefits of statins are due to their anti-inflammatory properties.76,283 These anti-inflammatory effects may make statins beneficial in the context of brain and neurological health as well.

Simvastatin efficiently crosses the blood-brain barrier and has been shown to exert potent anti-inflammatory and neuroprotective actions in the dopaminergic tract.77,78,284 In animal models, simvastatin was shown to attenuate the neurotoxicity of MPTP, which acts as a prodrug to the Parkinson’s disease-inducing neurotoxin MPP+. In fact, simvastatin accumulated in the substantia nigra and suppressed microglial activation, leading to reduced expression of inflammatory cytokines and increased dopaminergic neuroprotection.79 Another animal experiment found that simvastatin was able to completely reverse the decline in dopamine receptors associated with exposure to the neurotoxin 6-hydroxydopamine.80 Simvastatin was also shown to enhance the viability of neuroblastoma cells and neurons following treatment with MPP+ in vitro, suggesting a protective effect.285

In a large human clinical study involving over 700,000 subjects, use of simvastatin was associated with a 49% reduction in the likelihood of onset of Parkinson's symptoms, as well as a 54% reduction in the risk of dementia, suggesting a substantial neuroprotective effect.81

The neuroprotective effects of statins may also be due in part to improved blood flow to structures including the substantia nigra, as a result of reduced cerebrovascular atherosclerosis. In a clinical study involving 2,841 subjects over age 75 years at the beginning of the study, followed up over an average of almost six years, statin use at baseline was associated with a 16% reduced risk of developing parkinsonism, which for this study was defined as any combination of two or more Parkinsonian signs (movement abnormalities seen in Parkinson’s disease). Among study participants who died, decreased severity of atherosclerosis was found to explain 17% of the association between statins and parkinsonism. The authors believed the antioxidant and anti-inflammatory effects of statins could explain, in part, the remaining portion of the association. Statin use prior to death was associated with 37% reduced odds of atherosclerosis.324

Despite these findings, contrary results have since been observed in newer studies. A 2016 meta-analysis of 11 studies totaling over 3.5 million participants found individuals using statins showed a reduced risk of Parkinson’s disease; however, when analyzed individually, simvastatin’s association was not significant.286 These results were further supported by a systemic review and meta-analysis conducted in 2019 of 17 studies totaling nearly 4 million participants, which concluded that simvastatin use did not significantly reduce the risk of Parkinson’s disease, but atorvastatin (Lipitor) did.287 In fact, one retrospective case-control analysis of 2,322 patients with Parkinson’s disease found the use of lipophilic statins, such as simvastatin, was associated with an increased risk of Parkinson’s disease.288

As these studies were ongoing, simvastatin garnered interest as a potential therapeutic agent to slow progression of Parkinson’s disease. In a phase 2, double-blind, randomized, placebo-controlled clinical trial (PD STAT) initiated to examine the effect of simvastatin on Parkinson’s disease progression, 235 participants were allocated a low dose of simvastatin for 1 month (40 mg daily) followed by a high dose for 23 months (80 mg daily) or placebo. Simvastatin failed to slow the progression of Parkinson’s disease in phase II of this clinical trial. The trial was discontinued before entering phase III.289

Importantly, those taking any statin drug should be aware that statins deplete coenzyme Q10 (CoQ10) levels. If taking statins, supplement with CoQ10 and ensure maintenance of healthy CoQ10 blood levels by periodically having a CoQ10 blood test.

8 Diet

Low-Protein Diet/Protein Meal Redistribution

L-DOPA therapy is hindered by many obstacles, one of which is excess protein (specifically, aromatic amino acids) competing with L-DOPA for transport into the brain. Therefore, some studies have evaluated the effects of engaging in protein meal redistribution, involving eating dietary protein separate from dosing with L-DOPA.

Current research indicates that protein meal redistribution may be favorable with a low protein diet. It appears that protein meal redistribution reduces fluctuations, or "on-off periods" in response to L-DOPA therapy.121 Taking L-DOPA at least 30 minutes before consuming protein and/or having your highest protein meal at a time when L-DOPA is not needed may be an effective strategy. However, patients should speak with their physician to determine which dieting approach is appropriate for them.

Coffee Consumption

Coffee contains a multitude of pharmacologically active compounds, some of which have been shown to suppress oxidative stress and protect against diabetes, cancer, cognitive decline, and so on.122 Additionally, several epidemiological studies have found that those who consume large amounts of coffee are much less likely to develop Parkinson's disease.105,123,124

Coffee constituents (compounds) protect brain cells which can be extremely beneficial for Parkinson's disease patients. Coffee extracts have been shown to inhibit MAO-A and -B enzymes, a mechanism similar to that of some pharmaceutical Parkinson's therapies.125 Experimental models suggest that coffee constituents promote neuronal development and increase antioxidant defense systems in the brain.126,127

Green coffee extract contains more of the active antioxidant compounds than brewed coffee, and may be a promising option for Parkinson's disease patients.128 However, clinical trials have yet to confirm this potential benefit.

Intriguing research suggests that caffeine itself may be a potent anti-Parkinson agent. Upon ingestion, caffeine readily crosses the blood-brain barrier and blocks adenosine receptors, an effect responsible for many of its pharmacologic actions. The adenosine receptor system interacts with the dopaminergic system in several ways.129 Experimental studies have shown that caffeine binds to presynaptic adenosine receptors causing an increase in dopamine release, thereby temporarily ameliorating some symptoms of Parkinson's disease.130 In fact, some data from non-human primate studies indicate that adenosine receptor antagonists, like caffeine, may allow for a reduced dosage of L-DOPA. Data in mice also supports this notion, but more studies need to be done.131,132

In a clinical trial, a daily caffeine dose of 100 mg was shown to reduce "freezing." However, it appeared the subjects developed a tolerance after a few months. The researchers went on to suggest that caffeine might have therapeutic potential, but a periodic 2-week abstinence period may be required to maintain long term effectiveness.133

Current evidence suggests that coffee consumption may provide some neuroprotection and pharmacologic support, with very little potential downside for Parkinson's patients.

Flavonoid-Rich Diet

A healthy, well-balanced diet that provides adequate fruits and vegetables may be protective against Parkinson’s disease.314 A population-based study of 9,414 participants found that those who adhered to a Mediterranean diet, which emphasizes vegetables, fruits, olive oil, and whole grains, were less likely to develop Parkinson’s disease than those who had “unhealthy” diets.315 Plant foods provide numerous bioactive compounds, such as flavonoids, that may be beneficial in the context of Parkinson’s disease due to their neuroprotective activity.316 Various flavonoids have been shown to reduce the loss of dopaminergic neurons and improve behavioral symptoms in preclinical models of Parkinson’s disease.317-320 In addition, flavonoids may exert anti-inflammatory effects, improve mitochondrial function, and induce neurotrophic factors.321,322

A large cohort study that included 599 women from the Nurses’ Health Study and 652 men from the Health Professionals Follow-up Study who were diagnosed with Parkinson’s disease examined the relationship between flavonoid intake and mortality. Over 34 years of follow-up, the researchers found that risk of mortality was reduced by 47% for men in the highest quartile of flavonoid intake pre-diagnosis compared with men in the lowest quartile. In addition, greater flavonoid consumption post-diagnosis was associated with a 22% lower risk of mortality. The specific flavonoid subclasses anthocyanins, flavones, and flavan-3-ols, as well as flavonoid-rich foods like berries and red wine, were also protective when analyzed individually, with anthocyanins showing the greatest inverse association (37% and 47% risk reduction pre- and post-diagnosis, respectively).323

9 Nutrients

Conventional treatment of Parkinson's disease relies heavily on targeting amelioration of symptoms, without providing neuroprotection against continual cell death in the substantia nigra. On the other hand, a variety of natural ingredients have been shown to support neuronal health and promote mitochondrial function in a variety of ways, including suppressing oxidative stress and limiting inflammation. Many natural ingredients may have a complementary effect in combination with conventional therapies.

Coenzyme Q10 (CoQ10)

The connection between defects in mitochondrial energy management, oxidative stress, and neurodegeneration has led neuroscientists to explore several mitochondria-targeted supplemental compounds in the context of Parkinson’s disease. Coenzyme Q10 (CoQ10), also known as ubiquinone or ubiquinol because of its omnipresence in living cells, has garnered considerable interest in this area.134,135

CoQ10 is used in many biological reactions to facilitate the transport of electrons from energy-supplying nutrients and their metabolism within cells. Importantly, CoQ10 is essential for the generation of ATP, the most important source of energy in the body. CoQ10 deficiencies disrupt these reactions and may contribute to many age-related neurodegenerative conditions.290

Low plasma and platelet levels of CoQ10 have been observed in people with Parkinson's, suggesting a systemic deficiency state may be common in people with this disease. A 2008 study conducted in England observed, for the first time, reduced CoQ10 levels in cortical regions of the brain of Parkinson's disease patients.136 This deficiency, along with the role of oxidative stress in degeneration of dopaminergic neurons,291 supports the concept of utilizing CoQ10 supplementation to protect against oxidative stress and neurodegeneration in Parkinson’s disease. Moreover, several preclinical studies provided evidence that CoQ10 supplementation might mitigate the dopaminergic neurodegeneration characteristic of Parkinson’s disease.140,141,292 However, the protective effects observed in these preclinical studies were generally mild.293

Results from early clinical trials of CoQ10 in Parkinson’s disease were promising. In a multicenter clinical trial published in 2002, 80 treatment-naïve individuals with early Parkinson's disease were randomly assigned to receive either placebo or CoQ10 at daily doses of 300, 600, or 1,200 mg for 16 months or until disability required drug treatment. All subjects were scored using the standard Unified Parkinson Disease Rating Scale (UPDRS), for which higher scores indicate a progressively worsening disease state. The results were compelling with a mean change of 11.99 with placebo, 8.81 with the 300 mg dose, 10.82 with the 600 mg dose, and 6.69 with the 1,200 mg CoQ10 dose—a significant difference. All dosages were well tolerated. The authors concluded that "coenzyme Q10 appears to slow the progressive deterioration of function in Parkinson's disease."137 Two years later, the same researchers showed that dosages up to 3,000 mg/day of ubiquinone were safe and well tolerated, though plasma levels reached a plateau at 2,400 mg/day.138

Although these early clinical studies supported CoQ10 as a potential treatment for Parkinson’s disease, trial data obtained over recent years suggest less benefit than previously thought.

German researchers undertook a randomized controlled trial that was published in 2007. Unfortunately, their results were discouraging, showing no change in UPDRS scores. However, the subjects received a lower dose of CoQ10 (100 mg three times daily) over just three months. Unlike the previous trial, they also studied patients with "mid-range" Parkinson's disease, already requiring L-DOPA. Therefore, they would have been unable to detect meaningful neuroprotective effects that may be evident when CoQ10 supplementation is initiated preventively or in the very early stages of Parkinson’s disease. They did conclude, however, that CoQ10 was safe and well tolerated.139

Trial results published more recently have cast significant doubt on the potential of CoQ10 supplementation in providing clinical benefits in the context of Parkinson’s disease. A phase III, randomized, placebo-controlled, double-blind clinical trial conducted across 67 North American sites enrolled 600 participants 30 years of age or older who had been diagnosed with Parkinson’s disease within the previous five years. This study examined the effect of CoQ10 supplementation (1,200 mg or 2,400 mg daily) versus placebo on Parkinson’s disease severity over 16 months; results were published in 2014. The change in UPDRS score from baseline to final visit was used as the primary outcome to assess improvements following supplementation. CoQ10 supplementation was safe and well tolerated by participants, but the trial was halted as it showed no evidence of clinical benefit.294

These results showing lack of CoQ10 efficacy were affirmed in a 2016 meta-analysis of five randomized controlled trials totaling almost 1,000 participants. CoQ10 supplementation was compared with placebo in terms of motor functions and quality of life, as measured by UPDRS scores. UPDRS scores were not significantly different between CoQ10 and placebo, suggesting a lack of efficacy in providing symptomatic benefits for patients with Parkinson’s disease.295 A separate meta-analysis of eight randomized controlled trials totaling 899 patients with Parkinson’s disease further supported these findings. CoQ10 supplementation did not lead to significant differences in UPDRS scores compared with placebo but was well-tolerated by patients.296

Despite positive results from early clinical trials, supportive preclinical research, and strong biological plausibility for benefit, the totality of the clinical evidence available as of late 2021 suggests that CoQ10 supplementation may not provide as much benefit on the progression of Parkinson’s disease as previously hoped. Nevertheless, given the demonstrated tolerability of CoQ10 and the numerous benefits it has been shown to provide in clinical trials of other conditions, supplementation may still be reasonable for some people with Parkinson’s disease, or even those at a higher risk for developing the disease.297-299

Creatine

Creatine, an important amino acid-like compound, is vital to cellular energy management. Creatine deficiency is associated with neurological damage.142 Several animal studies have shown creatine, because of its "pro-mitochondrial" effect, to be effective in preventing or slowing the progression of Parkinson's disease.54,143,144 Influential Harvard neurologists noted that "creatine is a critical component in maintaining cellular energy homeostasis, and its administration has been reported to be neuroprotective in a wide number of both acute and chronic experimental models of neurological disease."145 Studies have shown that creatine is safe and well tolerated by patients with Parkinson's disease.146

In 2006, the Neuroprotective Exploratory Trials in Parkinson’s Disease (NET-PD) group at the National Institute of Neurological Disorders and Stroke (NINDS) studied 200 treatment-naïve subjects who had been diagnosed with Parkinson’s disease within the past five years. Subjects were randomly assigned to receive creatine 10 grams/day, the antibiotic drug minocycline (a proposed neuroprotectant) 200 mg/day, or placebo for 12 months while their scores on a standard Parkinson's disease rating scale were monitored. Both creatine and minocycline performed well, yet creatine showed a substantial edge in performance over minocycline. Tolerability of the treatment was 91% in the creatine group and 77% in the minocycline group.147 However, a follow-up study published by this same research group in 2015 failed to show a benefit associated with creatine supplementation (10 grams daily) for a minimum of five years.148

While it is unclear why the latter study did not identify a benefit with creatine supplementation, small differences in inclusion criteria and patient characteristics between the two studies may have contributed. Also, some evidence suggests that perhaps creatine in combination with other neuroprotective nutrients (as opposed to alone, as in the NET-PD studies) might be of benefit: a recent animal study found that creatine, in combination with CoQ10, conferred significant neuroprotection by reducing the accumulation of alpha-synuclein and suppressing lipid oxidation. In addition, animals being treated with the nutrient combination survived longer than those not being treated.149 Clinical trials are needed to assess the effects of creatine in combination with other neuroprotective nutrients in Parkinson’s patients.

Omega-3 Fatty Acids

These natural components of omega-3 fats, obtained chiefly from fish and some plant sources, exert significant anti-inflammatory action. Their concentration in nerve cell membranes decrease with age, oxidant stress, and in neurodegenerative disorders such as Parkinson's disease.150,151 In fact, researchers in Norway have presented convincing evidence of a systematic omega-3 deficit in Parkinson's disease, Alzheimer's disease, and autism, suggesting a fundamental neurological role for these vital fat molecules.152,153 Supplementation with the omega-3 DHA can favorably modify brain functions and has been proposed as a nutraceutical tool in Parkinson's and Alzheimer's disease.154

A study from Japan found that treatment of nerve cells with omega-3 prevents apoptosis, the programmed cell death that occurs in part as the result of inflammatory stimuli in the brain. Interestingly, results were a lot better when treatment was introduced before the chemical stresses that induced apoptosis were imposed, leading them to conclude that "dietary supplementation with [omega-3s] may be beneficial as a potential means to delay the onset of the diseases and/or their rate of progression."155

Canadian researchers took this study to the next level when they supplemented mice with omega-3 before injecting them with a Parkinson's inducing chemical.156 The mice were fed either a control or a high omega-3 diet for 10 months prior to injection. Control mice demonstrated a rapid loss of the dopamine producing cells in their substantia nigra accompanied by profound drops of dopamine levels in brain tissue. These effects were prevented in the mice receiving the high omega-3 diet.

A study of primates at the same institution demonstrated actual changes in Parkinson's symptoms, providing further compelling evidence for omega-3's protective and therapeutic effects. In this study, one group of animals was first treated for several months with L-DOPA before being given omega-3 DHA, while a second group was pre-treated with omega-3 DHA before starting on L-DOPA. The study was designed this way because L-DOPA, though effective in treating Parkinson's symptoms, as stated earlier in the protocol is also known to damage dopamine producing cells and induce dyskinesias. Omega-3 DHA reduced the occurrence of dyskinesias in both groups of monkeys, without altering the beneficial effects of L-DOPA. The researchers concluded that "DHA may represent a new approach to improve the quality of life of Parkinson's disease patients."157

B Vitamins

B vitamin deficiencies have long been implicated in many neurological disorders, including Parkinson's disease. Studies as early as the 1970's directed at demonstrating the effects of supplementation yielded discouraging results158-160 However, as our understanding of the close link between the toxic amino acid homocysteine and B vitamins grew, more targeted and mechanism-based studies became possible. Homocysteine levels are closely linked to folate, vitamins B6 and B12 status. Elevated homocysteine levels are found in cardiovascular disease as well as a variety of neurological and psychiatric disturbances.161-163 Also, L-DOPA treatment can itself lead to elevated homocysteine levels. As a result, more recent studies have led researchers to recommend B complex supplementation in those utilizing L-DOPA therapy.164

Definitive evidence supporting the benefit of this approach came from Singapore where Parkinson's disease patients, already on a stable dose of L-DOPA, were supplemented with pyridoxine (a common form of vitamin B6).165 Mean motor and activities of daily living scores improved significantly following supplementation, and worsened again when the supplements were stopped. Low serum folate is also found in Parkinson's disease patients, especially those taking L-DOPA.163 Canadian researchers demonstrated that a supplement containing folate and B12 could decrease plasma homocysteine levels in patients taking L-DOPA.166

A systematic review paper concluded that B vitamin supplementation may be of value for neurocognitive function.167 A similar review points to recent work with the active form of vitamin B6, pyridoxal-5' phosphate (P5P), noting that a number of neurological disorders including Parkinson's disease offer attractive therapeutic targets for this substance.168 The consensus among experts is that due to the deleterious effect that elevated homocysteine levels has on both Parkinson's itself and L-DOPA therapy, supplementation with folate, B6, and B12 is warranted.169-172

Thiamine, also known as vitamin B1, may benefit Parkinson’s patients. In 1999, low levels of free thiamine were detected in the cerebrospinal fluid of Parkinson’s disease patients.173 In 2013, researchers treated three newly diagnosed Parkinson’s patients with high-dose thiamine injections. Remarkably, the injections considerably improved the patients’ motor symptom deficits.174 Although only three subjects participated in this uncontrolled, informal clinical trial, the results corroborated very similar findings from another small trial in 2012.175

More recently, in 2015, an open-label clinical trial on 50 Parkinson’s disease patients showed that intramuscular injections of 100 mg of thiamine twice weekly led to significant and lasting improvements on a standardized Parkinson’s disease rating scale. Some participants—those with mild symptoms—had a complete clinical recovery. The benefits persisted throughout the follow-up period of up to about 2.2 years.176 In another open-label study, published in 2016, researchers treated 10 consecutive Parkinson’s patients with intramuscular 100 mg thiamine injections twice weekly without changing their medication regimens. Several measures of Parkinson’s disease symptoms improved significantly, and when the investigators increased the dosage of thiamine into the second month of treatment, the benefits became even more pronounced. Researchers speculated that the benefits of thiamine may arise from improvements in energy metabolism in surviving dopaminergic neurons in the substantia nigra.177 Additional clinical trials are needed to test whether oral thiamine, or thiamine derivatives such as benfotiamine, may have similarly beneficial effects.

Vitamin D

Vitamin D functions more like a hormone than a vitamin. Vitamin D receptors are expressed ubiquitously throughout the body, including on microglial cells.178 Upon activation by vitamin D, vitamin D receptors signal for increased or decreased expression of numerous genes, many of which are immunomodulatory.179

Several studies have shown that higher levels of vitamin D protect against the onset of Parkinson's disease symptoms. Also, that patients diagnosed with Parkinson's have lower serum vitamin D levels than those without the disease.180,181

Since many of the actions of vitamin D are anti-inflammatory, Life Extension believes that maintaining optimal vitamin D blood levels (50–80 ng/mL) may quell some of the inflammatory aspects of Parkinson's disease neurodegeneration. It is likely that having optimal vitamin D levels might decrease the activation of microglial cells and reduce the release of inflammatory cytokines.

Carnitine

Carnitine is a vital nutrient that serves as a co-factor in fatty acid metabolism. It helps to "ferry" large fat molecules into the mitochondrial "furnaces" where they are burned for energy, making it an important component of brain energy management and mitochondrial function.182 There is a growing body of literature suggesting that carnitine supplementation, through its support of brain energy management, protects against Parkinson's disease.

Mount Sinai researchers were able to prevent chemically induced Parkinson's disease in monkeys by pre-treating them with acetyl-l-carnitine, a readily absorbed form of the nutrient.183 Moreover, Italian researchers have studied carnitine as a neuroprotectant in the brains of methamphetamine users. Methamphetamines cause the same basic mitochondrial destruction and free radical brain damage as that seen in Parkinson's patients.182,184 This work has been extended in similar studies at the U.S. National Center for Toxicological Research.185

In an intriguing study, Chinese nutritional scientists in Shanghai explored in culture both acetyl-L-carnitine and lipoic acid (each alone and in combination with the other) in preventing Parkinson's disease-like changes in human neural cells. They found that both nutrients either alone or in combination, applied for four weeks prior to a Parkinson's disease-inducing chemical, protected the cells from mitochondrial dysfunction, oxidative damage, and an accumulation of the dangerous alpha-synuclein proteins. Notably, the combination of supplements was effective at 100- to 1000-fold lower concentrations than were required for either acting alone—powerful evidence that led the researchers to state that "this study provides important evidence that combining mitochondrial antioxidant/nutrients at optimal doses might be an effective and safe prevention strategy for Parkinson's disease."186

Green Tea

Increased tea consumption is correlated with reduced incidence of dementia, Alzheimer's and Parkinson's disease.187 Green tea contains valuable antioxidant polyphenols known to be protective against a host of chronic age-related conditions. There is tremendous scientific interest in green tea and its active compound epigallocatechin gallate (EGCG) as a neuroprotectant in Parkinson's disease; especially since when compared to many drugs, EGCG is extremely effective at penetrating brain tissue.188,189

Israeli researchers showed that they could prevent the cellular changes associated with Parkinson's by pre-treating mice with either green tea extracts or EGCG ahead of inducing the disease by chemical injection.188,190 This research has subsequently been repeated and extended in laboratories around the world.191-195 Utilizing the brain cell cultures pretreated to develop Parkinson's-like changes, the Israeli group also showed that green tea extracts prevented activation of the inflammation producing NF-kappaB system.190 EGCG's specific anti-inflammatory properties have been demonstrated to protect cultured brain tissue from the loss of dopaminergic cells as well.196 L-theanine, a component of green and black tea, was shown by Korean scientists to prevent dopaminergic cell death such as that seen in Parkinson's disease.197

Another potential benefit of green tea extract is its ability to inhibit the dopamine degrading enzyme COMT.74 This may help to sustain dopamine levels in ailing brain tissue thereby reducing the severity of symptoms.

Just as we use multiple combinations of prescription drugs to capitalize on their synergistic effects, we can capitalize on green tea's neuroprotective effects in Parkinson's and other neurodegenerative diseases.198 While more human studies are yet to be completed, green tea polyphenols have proven to exert powerful protection for dopaminergic neurons making them a key component in the prevention and treatment of Parkinson's disease.195,199-202

Resveratrol

Resveratrol is a polyphenolic antioxidant compound that has shown stunning potential in preventing cardiovascular disease and prolonging life.203-205 Not surprisingly, scientists interested in protecting brain tissue and enhancing the quality of life in aging individuals have directed their attention towards this remarkable compound.

Since dopamine itself is an oxidant compound which can contribute to the early destruction of neurons, Korean scientists studied the impact of resveratrol at preventing this paradoxical effect.206 They found that through the loss of mitochondrial function, human neural tissue treated with dopamine underwent rapid cell death. However, exposing the cells to resveratrol for one hour prior to dopamine treatment prevented cell loss and preserved mitochondrial function. In addition, Canadian scientists used resveratrol to prevent neuronal cell death caused by inflammation.207

Resveratrol's anti-inflammatory action was further explored by Chinese researchers who at first administered a Parkinson's disease-inducing chemical to rats, then gave them oral daily doses of resveratrol for 10 weeks. They found that after only two weeks of supplementation, the rats demonstrated significant improvement in their movement. Also, examination of their brains showed marked reduction in mitochondrial damage and loss of dopaminergic cells. Remarkably, they also found a reduction in the levels of COX-2 and TNF-alpha (inflammatory markers). They concluded with justifiable excitement that "resveratrol exerts a neuroprotective effect on [a chemically-] induced Parkinson's disease rat model, and this protection is related to the reduced inflammatory reaction."208

As with green tea extracts, it appears that resveratrol's potential for preventing Parkinson's disease may reside in its multi-modal mechanism of action targeting oxidant stress, and inflammation, for example.204 

Mucuna pruriensis a vine whose seeds contain a high concentration of naturally occurring L-DOPA and a variety of other psychoactive compounds.209 Compounds in Mucuna seeds act as AADC inhibitors, mimicking the action of carbidopa, and complementing the action L-DOPA in the central nervous system. In an animal experiment, Mucuna seed extract was shown to alleviate symptoms of chemically-induced Parkinson's with similar efficacy to traditional L-DOPA treatment, but without inducing dyskinesia.210 These results were repeated in another, similar trial.209

In a double-blind, randomized, placebo-controlled trial, Mucuna extract proved superior over standard L-DOPA/carbidopa therapy. Compared to traditional therapy, Mucuna led to a faster onset of symptom relief, longer duration of relief, and significantly fewer dyskinesias. The scientists conducting this study concluded that "The rapid onset of action and longer on time without concomitant increase in dyskinesias on mucuna seed powder formulation suggest that this natural source of L-dopa might possess advantages over conventional L-dopa preparations in the long term management of [Parkinson's disease]."211

Wild Green Oat Extract

Oats (Avena sativa L.) have been a foodstuff for many centuries and are an important cereal crop in North America, Europe, and Russia.212,213 Oats are the only known natural source of avenanthramides, which are bioavailable compounds with anti-inflammatory, anti-atherogenic, and antioxidant properties.214-216

Monoamine oxidase B (MAO-B) is responsible for the breakdown of dopamine in neurons217; however, excess MAO-B activity increases with age and depletes dopamine levels through excessive dopamine metabolism.218,219 Indeed, the loss of dopaminergic neurons is considered the hallmark of Parkinson’s disease, and restoration of dopaminergic signaling is the focus of most treatments.220-222 Drugs that inhibit MAO-B, such as deprenyl (Selegeline) and rasagiline (Azilect), are used in the treatment of early Parkinson’s disease.223-225 Blocking MAO-B with medications such as deprenyl not only raises dopamine levels in brain tissue, but also appears to be neuroprotective. MAO-B inhibitors block free radical formation that occurs during dopamine metabolism and blocks apoptosis (programmed cell death) of neurons.226

Wild green oat extract has shown the ability to inhibit MAO-B activity227,228 and demonstrated in numerous clinical trials to be a neuroprotective agent capable of improving cognitive function with as little as a single dose, while also increasing blood flow both systemically and to the brain by as much as 40%.229-232 Research demonstrates wild green oat extract is a neuroactive compound that shares a critical mechanism of action, MAO-B inhibition, with important Parkinson’s disease medications. This body of evidence suggests wild green oat extract is a promising natural therapy for those concerned with Parkinson’s disease.

Pyrroloquinoline Quinone (PQQ)

Pyrroloquinoline quinone, or PQQ, is a highly bioactive compound present in a vast range of cell types, and research suggests boosting PQQ levels may improve mitochondrial function, inhibit oxidative stress, and support neurological health.233-238 Since oxidative stress and mitochondrial dysfunction are believed to be key factors in Parkinson’s disease,239 PQQ is being actively studied as an agent to prevent and treat this condition.240

One mechanism by which PQQ may benefit those with Parkinson’s disease is by stimulating cerebral blood flow and oxygen use. Two studies using near-infrared spectrometry in healthy older adults have found supplementing with 20 mg PQQ daily for 12 weeks resulted in increased brain blood flow as well as increased oxygen utilization.237,241 In one of the studies, cognitive testing revealed PQQ was also associated with better preservation of cognitive function than placebo.237 These studies confirm earlier studies where PQQ prevented neurodegeneration and maintained memory function in a rodent experiment.242

A number of laboratory and animal studies have indicated PQQ can protect nerve cells from toxic and inflammatory damage by reducing oxidative stress and protecting mitochondria.240,243-245 Several preclinical studies have shown PQQ may prevent the accumulation of damaging amyloid proteins such as beta-amyloid,246-249 and might in this way protect against conditions such as Parkinson’s disease and Alzheimer’s disease.

Other Promising Nutrients

Curcumin. Curcumin, a derivative of the spice turmeric, through its potent modulation of the NF-kappa B system is a natural inhibitor of inflammation. It prevents chemically-induced changes in lab models of Parkinson's disease and exerts significant neuroprotection.250-257

Melatonin. The antioxidant hormone melatonin (synthesized and secreted by the pineal gland) may help to reduce the accumulation of alpha-synuclein proteins while preserving the cell's ability to make dopamine. It is also an invaluable sleep aid for Parkinson's patients, who often suffer from distressing problems with sleep.258-267

N-acetyl cysteine (NAC). NAC is a precursor to the potent cellular antioxidant glutathione. In animal models, NAC prevented dopamine induced neurotoxicity and protected against some of the damaging effects of alpha-synuclein proteins.268,269

Lipoic acid. Lipoic acid, a potent reducing agent, is considered a universal antioxidant due to its amphipathic nature (both fat- and water-soluble). Lipoic acid is produced naturally within the body and contributes to xenobiotic detoxification and antioxidant protection. It also contributes to cellular energy production.270 In addition to its ability to directly neutralize toxins and free radicals, lipoic acid bolsters levels of other cellular protectants such as glutathione and vitamin E.271

The low molecular weight of lipoic acid allows it to easily cross the blood-brain barrier, delivering neuroprotection within the central nervous system. Lipoic acid also combats inflammatory reactions.271 Large scale clinical trials have yet to be conducted in Parkinson's patients. However, given its potential for efficacy and excellent safety profile, lipoic acid should be considered as a therapeutic agent for Parkinson's disease.

Probiotics. Because dopaminergic signaling exerts considerable influence over intestinal function, constipation is a common problem in Parkinson's disease.

In a recent clinical trial, 40 Parkinson's patients complaining of constipation were treated with probiotics for five weeks. Probiotic therapy significantly increased the number of normal stools as well as reduced the incidence of bloating and abdominal pain.272

Phellodendron. The bark of the phellodendron tree (Phellodendron amurense), also known as the Amur cork tree, has been used for centuries in traditional Chinese medicine to treat various inflammatory conditions. An extract of phellodendron bark was found to reduce production of inflammatory cytokines and neurotoxic nitric oxide by microglia (specialized immune cells of the brain), indicating its potential usefulness in reducing the neuroinflammation and neurodegeneration underlying Parkinson’s disease.273 Phellodendron bark extract has also been shown to be a strong inhibitor of MAO-B, an enzyme that breaks down monoamine neurotransmitters, including dopamine.274,275

Adenosylcobalamin. Adenosylcobalamin is one of two biologically active forms of vitamin B12, the other being methylcobalamin. While methylcobalamin is a cofactor for enzymes that regulate methylation reactions, including homocysteine breakdown, adenosylcobalamin is needed for specialized enzyme pathways in the mitochondria.276,277 Abnormal patterns of genetic control of protein synthesis linked to neurological disorders such as Parkinson’s disease have been shown to be normalized when exposed to a high-vitamin B12 environment.278 In particular, adenosylcobalamin was found to regulate expression of the LRRK2 gene, prevent neurotoxicity, and restore function in dopamine-producing neurons in preclinical models of LRRK2 mutations.279 Because LRRK2 mutations are a common cause of familial Parkinson’s disease and increase the risk of sporadic Parkinson’s disease,280,281 LRRK2 inhibitors with actions such as those seen with adenosylcobalamin are being investigated as possible therapeutic agents for Parkinson’s disease.282

2023

  • Nov: Updated section on nicotine in Alternative and Emerging Therapies

2022

  • Apr: Updated section on simvastatin in Alternative and Emerging Therapies
  • Feb: Added section on flavonoids to Diet

2021

  • Nov: Added section on deep brain stimulation to Alternative and Emerging Therapies
  • Nov: Added section on focused ultrasound to Alternative and Emerging Therapies
  • Nov: Removed section on ablative surgery and deep-brain stimulation
  • Nov: Added section on simvastatin to Alternative and Emerging Therapies
  • Apr: Added section on phellodendron to Nutrients
  • Apr: Added section on adenosylcobalamin to Nutrients

2011

  • Sept: Comprehensive update & review

Disclaimer and Safety Information

This information (and any accompanying material) is not intended to replace the attention or advice of a physician or other qualified health care professional. Anyone who wishes to embark on any dietary, drug, exercise, or other lifestyle change intended to prevent or treat a specific disease or condition should first consult with and seek clearance from a physician or other qualified health care professional. Pregnant women in particular should seek the advice of a physician before using any protocol listed on this website. The protocols described on this website are for adults only, unless otherwise specified. Product labels may contain important safety information and the most recent product information provided by the product manufacturers should be carefully reviewed prior to use to verify the dose, administration, and contraindications. National, state, and local laws may vary regarding the use and application of many of the therapies discussed. The reader assumes the risk of any injuries. The authors and publishers, their affiliates and assigns are not liable for any injury and/or damage to persons arising from this protocol and expressly disclaim responsibility for any adverse effects resulting from the use of the information contained herein.

The protocols raise many issues that are subject to change as new data emerge. None of our suggested protocol regimens can guarantee health benefits. Life Extension has not performed independent verification of the data contained in the referenced materials, and expressly disclaims responsibility for any error in the literature.

  1. Parkinson's Disease Foundation. Statistics on Parkinson's. http://www.pdf.org/en/parkinson_statistics Accessed 8/1/2011.
  2. Heisters D. Parkinson's: symptoms, treatments and research. Br J Nurs. 2011 May 13-26;20(9):548-54.
  3. Parkinson J. An Essay on the Shaking Palsy. 1817. J Neuropsychiatry Clin Neurosci. 2002 Spring;14(2):223-36;discussion 222.
  4. Martinez TN and Greenamyre JT. Toxin Models of Mitochondrial Dysfunction in Parkinson's Disease. Antioxid Redox Signal. 2011 Jul 12. [Epub ahead of print]
  5. Astiz M et al. Effect of pesticides on cell survival in liver and brain rat tissues. Ecotoxicol Environ Saf. 2009 Oct;72(7):2025-32. Epub 2009 Jun 2.
  6. Fleming L et al. Parkinson's disease and brain levels of organochlorine pesticides. Ann Neurol. 1994 Jul;36(1):100-3.
  7. Betarbet R et al. Chronic systemic pesticide exposure reproduces features of Parkinson's disease. Nat Neurosci. 2000 Dec;3(12):1301-6.
  8. Brooks AI et al. Paraquat elicited neurobehavioral syndrome caused by dopaminergic neuron loss. Brain Res. 1999 Mar 27;823(1-2):1-10.
  9. Kenborg L et al. Parkinson's disease among gardeners exposed to pesticides - a Danish cohort study. Scand J Work Environ Health. 2011 Jun 17. pii: 3176. doi: 10.5271/sjweh.3176. [Epub ahead of print]
  10. Wang A et al. Parkinson's disease risk from ambient exposure to pesticides. Eur J Epidemiol. 2011 Jul;26(7):547-55. Epub 2011 Apr 20.
  11. Priyadarshi A et al. A meta-analysis of Parkinson's disease and exposure to pesticides. Neurotoxicology. 2000 Aug;21(4):435-40.
  12. Moretto A and Colosio C. Biochemical and toxicological evidence of neurological effects of pesticides: the example of Parkinson's disease. Neurotoxicology. 2011 Aug;32(4):383-91. Epub 2011 Mar 23.
  13. Corrigan FM et al. Organochlorine insecticides in substantia nigra in Parkinson's disease. J Toxicol Environ Health A. 2000 Feb 25;59(4):229-34.
  14. Kanthasamy AG et al. Dieldrin-induced neurotoxicity: relevance to Parkinson's disease pathogenesis. Neurotoxicology. 2005 Aug;26(4):701-19.
  15. Sun F et al. Dieldrin induces ubiquitin-proteasome dysfunction in alpha-synuclein overexpressing dopaminergic neuronal cells and enhances susceptibility to apoptotic cell death. J Pharmacol Exp Ther. 2005 Oct;315(1):69-79. Epub 2005 Jun 29.
  16. Steventon GB et al. A review of xenobiotic metabolism enzymes in Parkinson's disease and motor neuron disease. Drug Metabol Drug Interact. 2001;18(2):79-98.
  17. Williams A et al. Xenobiotic enzyme profiles and Parkinson's disease. Neurology. 1991 May;41(5 Suppl 2):29-32; discussion 32-3.
  18. Petrovitch H et al. Bowel movement frequency in late-life and substantia nigra neuron density at death. Mov Disord. 2009 Feb 15;24(3):371-6.
  19. Savica R et al. Medical records documentation of constipation preceding Parkinson disease: A case-control study. Neurology. 2009 Nov 24;73(21):1752-8.
  20. Ueki A et al. Life style risks of Parkinson's disease: association between decreased water intake and constipation. J Neurol. 2004 Oct;251 Suppl 7:vII18-23.
  21. Dickman MS. von Economo encephalitis. Arch Neurol. 2001 Oct;58(10):1696-8.
  22. Sardar AM et al. Dopamine deficits in the brain: the neurochemical basis of parkinsonian symptoms in AIDS. uroreport. 1996 Mar 22;7(4):910-2.
  23. PubMed Health. Parkinson's disease; Paralysis agitans; Shaking palsy. (http://www.ncbi.nlm.nih.gov/pubmedhealth/PMH0001762/). Accessed 8/17/2011.
  24. Mamo DC et al. Managing antipsychotic-induced parkinsonism. Drug Saf. 1999 Mar;20(3):269-75.
  25. Schouten HJ et al. Quality of Life of Elderly Patients With Antipsychotic-Induced Parkinsonism: A Cross-Sectional Study. J Am Med Dir Assoc. 2011 Feb 10.
  26. Bondon-Guitton E et al. Drug-induced parkinsonism: A review of 17 years' experience in a regional pharmacovigilance center in France. Mov Disord. 2011 Jun 14. doi: 10.1002/mds.23828. [Epub ahead of print]
  27. Francis PT. Biochemical and pathological correlates of cognitive and behavioural change in DLB/PDD. J Neurol. 2009 Aug;256 Suppl 3:280-5.
  28. Aarsland D and Kurz MW. The epidemiology of dementia associated with Parkinson disease. J Neurol Sci. 2010 Feb 15;289(1-2):18-22. Epub 2009 Sep 4.
  29. Francis PT et al. The cholinergic hypothesis of Alzheimer's disease: a review of progress. J Neurol Neurosurg Psychiatry. 1999 Feb;66(2):137-47.
  30. McKeith I. Commentary: DLB and PDD: the same or different? Is there a debate? Int Psychogeriatr. 2009 Apr;21(2):220-4. Epub 2009 Jan 28.
  31. Samii A et al. Parkinson's disease. Lancet. 2004 May 29;363(9423):1783-93.
  32. Lesage S and Brice A. Parkinson's disease: from monogenic forms to genetic susceptibility factors. Hum Mol Genet. 2009 Apr 15;18(R1):R48-59.
  33. Mata IF et al. SNCA variant associated with Parkinson disease and plasma alpha-synuclein level. Arch Neurol. 2010 Nov;67(11):1350-6.
  34. Pihlstrom L et al. Genetic variability in SNCA and Parkinson's disease. Neurogenetics. 2011 Jul 29. [Epub ahead of print]
  35. Lewis KA et al. Abnormal neurites containing C-terminally truncated alpha-synuclein are present in Alzheimer's disease without conventional Lewy body pathology. Am J Pathol. 2010 Dec;177(6):3037-50. Epub 2010 Nov 5.
  36. Singleton AB et al. alpha-Synuclein locus triplication causes Parkinson's disease. Science. 2003 Oct 31;302(5646):841.
  37. Papapetropoulos S et al. Clinical heterogeneity of the LRRK2 G2019S mutation. Arch Neurol. 2006 Sep;63(9):1242-6.
  38. Poorkaj P et al. parkin mutation analysis in clinic patients with early-onset Parkinson [corrected] disease. Am J Med Genet A. 2004 Aug 15;129A(1):44-50.
  39. Yu W et al. The PINK1/Parkin pathway regulates mitochondrial dynamics and function in mammalian hippocampal and dopaminergic neurons. Hum Mol Genet. 2011 Aug 15;20(16):3227-40. Epub 2011 May 25.
  40. Whitworth AJ and Pallanck LJ. The PINK1/Parkin pathway: a mitochondrial quality control system? J Bioenerg Biomembr. 2009 Dec;41(6):499-503.
  41. Khan NL et al. Clinical and subclinical dopaminergic dysfunction in PARK6-linked parkinsonism: an 18F-dopa PET study. Ann Neurol. 2002 Dec;52(6):849-53.
  42. Zhang X et al. Mutation analysis of PINK1 gene in patients with early-onset Parkinsonism. Zhong Nan Da Xue Xue Bao Yi Xue Ban. 2011 Jun;36(6):490-7.
  43. Xu X et al. The familial Parkinson's disease gene DJ-1 (PARK7) is expressed in red cells and plays a role in protection against oxidative damage. Blood Cells Mol Dis. 2010 Oct;45(3):227-32.
  44. van Duijn CM et al. Park7, a novel locus for autosomal recessive early-onset parkinsonism, on chromosome 1p36. Am J Hum Genet. 2001 Sep;69(3):629-34. Epub 2001 Jul 2.
  45. Bonifati V et al. mal recessive early onset parkinsonism is linked to three loci: PARK2, PARK6, and PARK7. Neurol Sci. 2002 Sep;23 Suppl 2:S59-60.
  46. Hering R et al. Novel homozygous p.E64D mutation in DJ1 in early onset Parkinson disease (PARK7). Hum Mutat. 2004 Oct;24(4):321-9.
  47. Inden M, Taira T, Kitamura Y, et al. PARK7 DJ-1 protects against degeneration of nigral dopaminergic neurons in Parkinson’s disease rat model. Neurobiol Dis. 2006 Oct;24(1):144-58.
  48. Hampshire DJ et al. Kufor-Rakeb syndrome, pallido-pyramidal degeneration with supranuclear upgaze paresis and dementia, maps to 1p36. J Med Genet. 2001 Oct;38(10):680-2.
  49. Ugolino J et al. Mutant Atp13a2 proteins involved in parkinsonism are degraded by ER-associated degradation and sensitize cells to ER-stress induced cell death. Hum Mol Genet. 2011 Jun 23. [Epub ahead of print].
  50. Park JS et al. Pathogenic effects of novel mutations in the P-type ATPase ATP13A2 (PARK9) causing Kufor-Rakeb syndrome, a form of early-onset parkinsonism. Hum Mutat. 2011  Aug;32(8):956-64. doi: 10.1002/humu.21527. Epub 2011 Jul 12.
  51. Fong CY et al. Juvenile parkinsonism associated with heterozygous frameshift ATP13A2 gene mutation. Eur J Paediatr Neurol. 2011 May;15(3):271-5. Epub 2011 Feb 12.
  52. Kempster PA et al. Relationships between age and late progression of Parkinson's disease: a clinico-pathological study. Brain. 2010 Jun;133(Pt 6):1755-62. Epub 2010 Apr 5.
  53. Lee MS et al. The effect of age on motor deficits and cerebral glucose metabolism of Parkinson's disease. Acta Neurol Scand. 2011 Sep;124(3):196-201. doi: 10.1111/j.1600-0404.2010.01446.x. Epub 2010 Sep 29.
  54. Schapira AH. Mitochondria in the aetiology and pathogenesis of Parkinson's disease. Lancet Neurol. 2008 Jan;7(1):97-109.
  55. Zhu J et al. Mitochondrial dysfunction in Parkinson's disease. J Alzheimers Dis. 2010;20 Suppl 2:S325-34.
  56. Lin TK et al. Mitochondrial dysfunction and biogenesis in the pathogenesis of Parkinson's disease. Chang Gung Med J. 2009 Nov-Dec;32(6):589-99.
  57. Van Humbeeck et al. Parkin interacts with ambra1 to induce mitophagy. J Neurosci. 2011 Jul 13;31(28):10249-61.
  58. Geisler S et al. PINK1/Parkin-mediated mitophagy is dependent on VDAC1 and p62/SQSTM1. Nat Cell Biol. 2010 Feb;12(2):119-31. Epub 2010 Jan 24.
  59. Song DD et al. Enhanced substantia nigra mitochondrial pathology in human alpha-synuclein transgenic mice after treatment with MPTP. Exp Neurol. 2004 Apr;186(2):158-72.
  60. Kraytsberg Y et al. Mitochondrial DNA deletions are abundant and cause functional impairment in aged human substantia nigra neurons. Nat Genet. 2006 May;38(5):518-20. Epub 2006 Apr 9.
  61. de Castro IP et al. Mitochondrial quality control and Parkinson's disease: a pathway unfolds. Mol Neurobiol. 2011 Apr;43(2):80-6. Epub 2010 Dec 1.
  62. Gautam AH and Zeevalk GD. Characterization of reduced and oxidized dopamine and 3,4-dihydrophenylacetic acid, on brain mitochondrial electron transport chain activities. Biochim Biophys Acta. 2011 Jul;1807(7):819-28. Epub 2011 Apr 2.
  63. Khan FH et al. Inhibition of rat brain mitochondrial electron transport chain activity by dopamine oxidation products during extended in vitro incubation: implications for Parkinson's disease. Biochim Biophys Acta. 2005 Jun 30;1741(1-2):65-74. Epub 2005 Apr 14.
  64. Mosharov EV. Interplay between cytosolic dopamine, calcium, and alpha-synuclein causes selective death of substantia nigra neurons. Neuron. 2009 Apr 30;62(2):218-29.
  65. Chu CT. Diversity in the regulation of autophagy and mitophagy: lessons from Parkinson's disease. Parkinsons Dis. 2011;2011:789431. Epub 2011 Mar 17.
  66. Li WW et al. Localization of alpha-synuclein to mitochondria within midbrain of mice. Neuroreport. 2007 Oct 8;18(15):1543-6.
  67. Liu G et al. alpha-Synuclein is differentially expressed in mitochondria from different rat brain regions and dose-dependently down-regulates complex I activity. Neurosci Lett. 2009 May 1;454(3):187-92. Epub 2009 Feb 28.
  68. Devi L et al. Mitochondrial import and accumulation of alpha-synuclein impair complex I in human dopaminergic neuronal cultures and Parkinson disease brain. J Biol Chem. 2008 Apr 4;283(14):9089-100. Epub 2008 Feb 1.
  69. Iseki E et al. Degeneration process of Lewy bodies in the brains of patients with dementia with Lewy bodies using alpha-synuclein-immunohistochemistry. Neurosci Lett. 2000 May 26;286(1):69-73.
  70. McGeer PL et al. Reactive microglia are positive for HLA-DR in the substantia nigra of Parkinson's and Alzheimer's disease brains. Neurology. 1988 Aug;38(8):1285-91.
  71. McGeer PL et al. Inflammation and neurodegeneration in Parkinson's disease. Parkinsonism Relat Disord. 2004 May;10 Suppl 1:S3-7.
  72. Barcia C et al. Inflammatory response in Parkinsonism. J Neural Transm Suppl. 2009;(73):245-52.
  73. Glass CK et al. Mechanisms underlying inflammation in neurodegeneration. Cell. 2010 Mar 19;140(6):918-34.
  74. Chen H et al. Nonsteroidal antiinflammatory drug use and the risk for Parkinson's disease. Ann Neurol. 2005 Dec;58(6):963-7.
  75. Driver JA et al. Use of non-steroidal anti-inflammatory drugs and risk of Parkinson's disease: nested case-control study. BMJ. 2011 Jan 20;342:d198. doi: 10.1136/bmj.d198.
  76. Quist-Paulsen P. Statins and inflammation: an update. Curr Opin Cardiol. 2010 Jul;25(4):399-405.
  77. Roy A et al. Prospects of statins in Parkinson disease. Neuroscientist. 2011 Jun;17(3):244-55. Epub 2011 Jan 20.
  78. Yan J et al. Simvastatin prevents dopaminergic neurodegeneration in experimental parkinsonian models: the association with anti-inflammatory responses. PLoS One. 2011;6(6):e20945. Epub 2011 Jun 22.
  79. Ghosh A et al. Simvastatin inhibits the activation of p21ras and prevents the loss of dopaminergic neurons in a mouse model of Parkinson's disease. J Neurosci. 2009 Oct 28;29(43):13543-56.
  80. Wang Q et al. Simvastatin reverses the downregulation of dopamine D1 and D2 receptor expression in the prefrontal cortex of 6-hydroxydopamine-induced Parkinsonian rats. Brain Res. 2005 May 31;1045(1-2):229-33. Epub 2005 Apr 22.
  81. Wolozin B et al. Simvastatin is associated with a reduced incidence of dementia and Parkinson's disease. BMC Med. 2007 Jul 19;5:20.
  82. Rice JE et al. Disordered respiration as a levodopa-induced dyskinesia in Parkinson's disease. Mov Disord. 2002 May;17(3):524-7.
  83. Jenner P et al. Molecular mechanisms of L-DOPA-induced dyskinesia. ture Reviews Neuroscience9, 665-677 (September 2008).
  84. Rajagopalan P et al. Homocysteine effects on brain volumes mapped in 732 elderly individuals. Neuroreport. 2011 Jun 11;22(8):391-5.
  85. Kloppenborg RP et al. Homocysteine and cerebral small vessel disease in patients with symptomatic atherosclerotic disease. The SMART-MR study. Atherosclerosis. 2011 Jun;216(2):461-6. Epub 2011 Feb 24.
  86. Zoccolella S et al. Hyperhomocysteinemia in L-dopa treated patients with Parkinson's disease: potential implications in cognitive dysfunction and dementia? Curr Med Chem. 2010;17(28):3253-61.
  87. Corona T et al. A longitudinal study of the effects of an L-dopa drug holiday on the course of Parkinson's disease. Clin Neuropharmacol. 1995 Aug;18(4):325-32.
  88. Koziorowski D and Friedman A et al. Levodopa "drug holiday" with amantadine infusions as a treatment of complications in Parkinson's disease. Mov Disord. 2007 May 15;22(7):1033-6.
  89. Antonini A et al. A reassessment of risks and benefits of dopamine agonists in Parkinson's disease. Lancet Neurol. 2009 Oct;8(10):929-37. Epub 2009 Aug 24.
  90. Katzenschlager R et al. Fourteen-year final report of the randomized PDRG-UK trial comparing three initial treatments in PD. Neurology. 2008 Aug 12;71(7):474-80. Epub 2008 Jun 25.
  91. Mizuno Y et al. Early addition of selegiline to L-Dopa treatment is beneficial for patients with Parkinson disease. Clin Neuropharmacol. 2010 Jan-Feb;33(1):1-4.
  92. Zhao YJ et al. Selegiline use is associated with a slower progression in early Parkinson's disease as evaluated by Hoehn and Yahr Stage transition times. Parkinsonism Relat Disord. 2011 Mar;17(3):194-7. Epub 2010 Dec 14.
  93. Palhagen S et al. Selegiline slows the progression of the symptoms of Parkinson disease. Neurology. 2006 Apr 25;66(8):1200-6. Epub 2006 Mar 15.
  94. Imamura K et al. The relationship between depression and regional cerebral blood flow in Parkinson's disease and the effect of selegiline treatment. Acta Neurol Scand. 2011 Jul;124(1):28-39. Epub 2010 Sep 29.
  95. Braga CA et al. The anti-Parkinsonian drug selegiline delays the nucleation phase of α-synuclein aggregation leading to the formation of nontoxic species. J Mol Biol. 2011 Jan 7;405(1):254-73. Epub 2010 Nov 2.
  96. Dimpfel W et al. Effects of rasagiline, its metabolite aminoindan and selegiline on glutamate receptor mediated signalling in the rat hippocampus slice in vitro. BMC Pharmacol. 2011 Feb 21;11:2.
  97. Rascol O et al. A double-blind, delayed-start trial of rasagiline in Parkinson's disease (the ADAGIO study): prespecified and post-hoc analyses of the need for additional therapies, changes in UPDRS scores, and non-motor outcomes. Lancet Neurol. 2011 May;10(5):415-23. Epub 2011 Apr 7.
  98. Sawada H et al. Amantadine for dyskinesias in Parkinson's disease: a randomized controlled trial. PLoS One. 2010 Dec 31;5(12):e15298.
  99. Thomas A et al. Duration of amantadine benefit on dyskinesia of severe Parkinson's disease. J Neurol Neurosurg Psychiatry. 2004 Jan;75(1):141-3.
  100. Exley R and Cragg SJ. Presynaptic nicotinic receptors: a dynamic and diverse cholinergic filter of striatal dopamine neurotransmission. Br J Pharmacol. 2008 Mar;153 Suppl 1:S283-97. Epub 2007 Nov 26.
  101. Pimlott SL et al. Nicotinic acetylcholine receptor distribution in Alzheimer's disease, dementia with Lewy bodies, Parkinson's disease, and vascular dementia: in vitro binding study using 5-[(125)i]-a-85380. Neuropsychopharmacology. 2004 Jan;29(1):108-16.
  102. Shimohama S. Nicotinic receptor-mediated neuroprotection in neurodegenerative disease models. Biol Pharm Bull. 2009 Mar;32(3):332-6.
  103. Hong DP et al. Smoking and Parkinson's disease: does nicotine affect alpha-synuclein fibrillation? Biochim Biophys Acta. 2009 Feb;1794(2):282-90. Epub 2008 Oct 25.
  104. Tanaka K et al. Active and passive smoking and risk of Parkinson's disease. Acta Neurol Scand. 2010 Dec;122(6):377-82.
  105. Tan EK et al. Dose-dependent protective effect of coffee, tea, and smoking in Parkinson's disease: a study in ethnic Chinese. J Neurol Sci. 2003 Dec 15;216(1):163-7.
  106. Holms AD et al. Acute nicotine enhances strategy-based semantic processing in Parkinson's disease. Int J Neuropsychopharmacol. 2011 Aug;14(7):877-85. Epub 2011 Feb 1.
  107. Quik M et al. Nicotine and Parkinson's disease: implications for therapy. Mov Disord. 2008 Sep 15;23(12):1641-52.
  108. Xiao BG et al. Cell biology and clinical promise of G-CSF: immunomodulation and neuroprotection. J Cell Mol Med. 2007 Nov-Dec;11(6):1272-90.
  109. Khatibi NH et al. Granulocyte colony-stimulating factor treatment provides neuroprotection in surgically induced brain injured mice. Acta Neurochir Suppl. 2011;111:265-9.
  110. Song S et al. Granulocyte-colony stimulating factor (G-CSF) enhances recovery in mouse model of Parkinson's disease. Neurosci Lett. 2011 Jan 7;487(2):153-7. Epub 2010 Oct 14.
  111. McCollum M et al. Post-MPTP treatment with granulocyte colony-stimulating factor improves nigrostriatal function in the mouse model of Parkinson's disease. Mol Neurobiol. 2010 Jun;41(2-3):410-9. Epub 2010 Apr 21.
  112. Sanchez-Ramos J et al. Granulocyte colony stimulating factor decreases brain amyloid burden and reverses cognitive impairment in Alzheimer's mice. Neuroscience. 2009 Sep 29;163(1):55-72. Epub 2009 Jun 14.
  113. Freed CR et al. Transplantation of embryonic dopamine neurons for severe Parkinson's disease. N Engl J Med. 2001 Mar 8;344(10):710-9.
  114. Kim HJ. Stem cell potential in Parkinson's disease and molecular factors for the generation of dopamine neurons. Biochim Biophys Acta. 2011 Jan;1812(1):1-11. Epub 2010 Aug 14.
  115. Dobkin RD et al. Cognitive-Behavioral Therapy for Depression in Parkinson's Disease: A Randomized, Controlled Trial. Am J Psychiatry. 2011 Jun 15. [Epub ahead of print]
  116. Vaughan CP et al. Behavioral therapy to treat urinary incontinence in Parkinson disease. Neurology. 2011 May 10;76(19):1631-4.
  117. Allen NE et al. Balance and falls in Parkinson's disease: A meta-analysis of the effect of exercise and motor training. Mov Disord. 2011 Aug 1;26(9):1605-15. doi: 10.1002/mds.23790. Epub 2011 Jun 14.
  118. Jak AJ. The Impact of Physical and Mental Activity on Cognitive Aging. Curr Top Behav Neurosci. 2011 Aug 5. [Epub ahead of print]
  119. Lau YS et al. Neuroprotective effects and mechanisms of exercise in a chronic mouse model of Parkinson's disease with moderate neurodegeneration. Eur J Neurosci. 2011 Apr;33(7):1264-74. doi: 10.1111/j.1460-9568.2011.07626.x. Epub 2011 Mar 7.
  120. Cereda E et al. Low-protein and protein-redistribution diets for Parkinson's disease patients with motor fluctuations: a systematic review. Mov Disord. 2010 Oct 15;25(13):2021-34.
  121. Butt MS and Sultan MT. Coffee and its consumption: benefits and risks. Crit Rev Food Sci Nutr. 2011 Apr;51(4):363-73.
  122. Hu G et al. Coffee and tea consumption and the risk of Parkinson's disease. Mov Disord. 2007 Nov 15;22(15):2242-8.
  123. Saaksjarvi K et al. Prospective study of coffee consumption and risk of Parkinson's disease. Eur J Clin Nutr. 2008 Jul;62(7):908-15. Epub 2007 May 16.
  124. Herraiz T and Chaparro C. Human monoamine oxidase enzyme inhibition by coffee and beta-carbolines norharman and harman isolated from coffee. Life Sci. 2006 Jan 18;78(8):795-802. Epub 2005 Aug 31.
  125. Abreu RV et al. Chronic coffee and caffeine ingestion effects on the cognitive function and antioxidant system of rat brains. Pharmacol Biochem Behav. 2011 Oct;99(4):659-64. Epub 2011 Jun 15.
  126. Tohda C et al. Trigonelline-induced neurite outgrowth in human neuroblastoma SK-N-SH cells. Biol Pharm Bull. 1999 Jul;22(7):679-82.
  127. Farah A et al. Chlorogenic acids from green coffee extract are highly bioavailable in humans. J Nutr. 2008 Dec;138(12):2309-15.
  128. Xie X et al. Adenosine and dopamine receptor interactions in striatum and caffeine-induced behavioral activation. Comp Med. 2007 Dec;57(6):538-45.
  129. Hauser RA et al. Adenosine A2A receptor antagonists for Parkinson's disease: rationale, therapeutic potential and clinical experience. Drugs Aging. 2005;22(6):471-82.
  130. Kanda T et al. Combined use of the adenosine A(2A) antagonist KW-6002 with L-DOPA or with selective D1 or D2 dopamine agonists increases antiparkinsonian activity but not dyskinesia in MPTP-treated monkeys. Exp Neurol. 2000 Apr;162(2):321-7.
  131. Matsuya T et al. Synergistic effects of adenosine A2A antagonist and L-DOPA on rotational behaviors in 6-hydroxydopamine-induced hemi-Parkinsonian mouse model. J Pharmacol Sci. 2007 Mar;103(3):329-32. Epub 2007 Mar 7.
  132. Kitagawa M et al. ffects of caffeine on the freezing of gait in Parkinson's disease. Mov Disord. 2007 Apr 15;22(5):710-2.
  133. Dhanasekaran M, Karuppagounder SS, Uthayathas S, et al. Effect of dopaminergic neurotoxin MPTP/MPP+ on coenzyme Q content. Life Sci. Jul 18 2008;83(3-4):92-95.
  134. Henchcliffe C, Beal MF. Mitochondrial biology and oxidative stress in Parkinson disease pathogenesis. Nat Clin Pract Neurol. Nov 2008;4(11):600-609.
  135. Hargreaves IP, Lane A, Sleiman PM. The coenzyme Q10 status of the brain regions of Parkinson's disease patients. Neurosci Lett. Dec 5 2008;447(1):17-19.
  136. Shults CW, Oakes D, Kieburtz K, et al. Effects of coenzyme Q10 in early Parkinson disease: evidence of slowing of the functional decline. Arch Neurol. Oct 2002;59(10):1541-1550.
  137. Shults CW, Flint Beal M, Song D, Fontaine D. Pilot trial of high dosages of coenzyme Q10 in patients with Parkinson's disease. Exp Neurol. Aug 2004;188(2):491-494.
  138. Storch A et al. Randomized, double-blind, placebo-controlled trial on symptomatic effects of coenzyme Q(10) in Parkinson disease. Arch Neurol. 2007 Jul;64(7):938-44. Epub 2007 May 14.
  139. Abdin AA, Hamouda HE. Mechanism of the neuroprotective role of coenzyme Q10 with or without L-dopa in rotenone-induced parkinsonism. Neuropharmacology. Dec 2008;55(8):1340-1346.
  140. Cleren C, Yang L, Lorenzo B, et al. Therapeutic effects of coenzyme Q10 (CoQ10) and reduced CoQ10 in the MPTP model of Parkinsonism. J Neurochem. Mar 2008;104(6):1613-1621.
  141. Wyss M, Schulze A. Health implications of creatine: can oral creatine supplementation protect against neurological and atherosclerotic disease? Neuroscience. 2002;112(2):243-260.
  142. Beal MF. Bioenergetic approaches for neuroprotection in Parkinson's disease. Ann Neurol. 2003;53 Suppl 3:S39-47; discussion S47-38.
  143. Fernandez-Espejo E. Pathogenesis of Parkinson's disease: prospects of neuroprotective and restorative therapies. Mol Neurobiol. Feb 2004;29(1):15-30.
  144. Klein AM, Ferrante RJ. The neuroprotective role of creatine. Subcell Biochem. 2007;46:205-243.
  145. Bender A et al. Long-term creatine supplementation is safe in aged patients with Parkinson disease. Nutr Res. 2008 Mar;28(3):172-8.
  146. NINDS NET-PD Investigators. A randomized, double-blind, futility clinical trial of creatine and minocycline in early Parkinson disease. Neurology. Mar 14 2006;66(5):664-671.
  147. Writing Group for the NETiPDI. Effect of creatine monohydrate on clinical progression in patients with parkinson disease: A randomized clinical trial. JAMA : the journal of the American Medical Association. 2015;313(6):584-593.
  148. Yang L et al. Combination therapy with coenzyme Q10 and creatine produces additive neuroprotective effects in models of Parkinson's and Huntington's diseases. J Neurochem. 2009 Jun;109(5):1427-39. Epub 2009 Mar 28.
  149. Youdim KA, Martin A, Joseph JA. Essential fatty acids and the brain: possible health implications. Int J Dev Neurosci. Jul-Aug 2000;18(4-5):383-399.
  150. Montine KS, Quinn JF, Zhang J, et al. Isoprostanes and related products of lipid peroxidation in neurodegenerative diseases. Chem Phys Lipids. Mar 2004;128(1-2):117-124.
  151. Saugstad LF. Infantile autism: a chronic psychosis since infancy due to synaptic pruning of the supplementary motor area. Nutr Health. 2008;19(4):307-317.
  152. Saugstad LF. Are neurodegenerative disorder and psychotic manifestations avoidable brain dysfunctions with adequate dietary omega-3? Nutr Health. 2006;18(2):89-101.
  153. Calon F, Cole G. Neuroprotective action of omega-3 polyunsaturated fatty acids against neurodegenerative diseases: evidence from animal studies. Prostaglandins Leukot Essent Fatty Acids. Nov-Dec 2007;77(5-6):287-293.
  154. Wu Y, Tada M, Takahata K, Tomizawa K, Matsui H. Inhibitory effect of polyunsaturated fatty acids on apoptosis induced by etoposide, okadaic acid and AraC in Neuro2a cells. Acta Med Okayama. Jun 2007;61(3):147-152.
  155. Bousquet M, Saint-Pierre M, Julien C, Salem N, Jr., Cicchetti F, Calon F. Beneficial effects of dietary omega-3 polyunsaturated fatty acid on toxin-induced neuronal degeneration in an animal model of Parkinson's disease. FASEB J. Apr 2008;22(4):1213-1225.
  156. Samadi P, Gregoire L, Rouillard C, Bedard PJ, Di Paolo T, Levesque D. Docosahexaenoic acid reduces levodopa-induced dyskinesias in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine monkeys. Ann Neurol. Feb 2006;59(2):282-288.
  157. Yahr MD, Duvoisin RC, Cote L, Cohen G. Pyridoxine, DOPA, and Parkinsonism. Adv Biochem Psychopharmacol. 1972;4:185-194.
  158. McGeer PL, Zeldowicz L, McGeer EG. A clinical trial of folic acid in Parkinson's disease. Can Med Assoc J. Jan 22 1972;106(2):145-146 passim.
  159. Schwarz J, Trenkwalder C, Gasser T, Arnold G, Oertel WH. Folinic acid therapy fails to improve early Parkinson's disease: a two week placebo controlled clinical trial. J Neural Transm Park Dis Dement Sect. 1992;4(1):35-41.
  160. Bottiglieri T, Hyland K, Reynolds EH. The clinical potential of ademetionine (S-adenosylmethionine) in neurological disorders. Drugs. Aug 1994;48(2):137-152.
  161. Martignoni E, Tassorelli C, Nappi G, Zangaglia R, Pacchetti C, Blandini F. Homocysteine and Parkinson's disease: a dangerous liaison? J Neurol Sci. Jun 15 2007;257(1-2):31-37.
  162. Obeid R, McCaddon A, Herrmann W. The role of hyperhomocysteinemia and B-vitamin deficiency in neurological and psychiatric diseases. Clin Chem Lab Med. 2007;45(12):1590-1606.
  163. Siniscalchi A, Mancuso F, Gallelli L, Ferreri Ibbadu G, Biagio Mercuri N, De Sarro G. Increase in plasma homocysteine levels induced by drug treatments in neurologic patients. Pharmacol Res. Nov 2005;52(5):367-375.
  164. Tan EK, Cheah SY, Fook-Chong S, et al. Functional COMT variant predicts response to high dose pyridoxine in Parkinson's disease. Am J Med Genet B Neuropsychiatr Genet. Aug 5 2005;137B(1):1-4.
  165. Postuma RB, Espay AJ, Zadikoff C, et al. Vitamins and entacapone in levodopa-induced hyperhomocysteinemia: a randomized controlled study. Neurology. Jun 27 2006;66(12):1941-1943.
  166. Balk E, Chung M, Raman G, et al. B vitamins and berries and age-related neurodegenerative disorders. Evid Rep Technol Assess (Full Rep). Apr 2006(134):1-161.
  167. Amadasi A, Bertoldi M, Contestabile R, et al. Pyridoxal 5'-phosphate enzymes as targets for therapeutic agents. Curr Med Chem. 2007;14(12):1291-1324.
  168. Zoccolella S, Iliceto G, deMari M, Livrea P, Lamberti P. Management of L-Dopa related hyperhomocysteinemia: catechol-O-methyltransferase (COMT) inhibitors or B vitamins? Results from a review. Clin Chem Lab Med. 2007;45(12):1607-1613.
  169. Qureshi  GA, Qureshi AA, Devrajani BR, Chippa MA, Syed SA. Is the deficiency of vitamin B12 related to oxidative stress and neurotoxicity in Parkinson's patients? CNS Neurol Disord Drug Targets. Feb 2008;7(1):20-27.
  170. Muller T. Role of homocysteine in the treatment of Parkinson's disease. Expert Rev Neurother. Jun 2008;8(6):957-967.
  171. Dos Santos EF, Busanello EN, Miglioranza A, et al. Evidence that folic acid deficiency is a major determinant of hyperhomocysteinemia in Parkinson s disease. Metab Brain Dis. Mar 18 2009.
  172. Jimenez-Jimenez FJ, Molina JA, Hernanz A, Fernandez-Vivancos E, de Bustos F, Barcenilla B, . . . Villanueva C. Cerebrospinal fluid levels of thiamine in patients with Parkinson's disease. Neuroscience letters.Aug 13 1999;271(1):33-36.
  173. Costantini A, Pala MI, Compagnoni L, Colangeli M. High-dose thiamine as initial treatment for Parkinson's disease. BMJ case reports.Aug 28 2013;2013.
  174. Luong Kvq, Nguyen LTH. The Beneficial Role of Thiamine in Parkinson’s Disease: Preliminary Report. 2012.
  175. Costantini A, Pala MI, Grossi E, Mondonico S, Cardelli LE, Jenner C, . . . Fancellu R. Long-Term Treatment with High-Dose Thiamine in Parkinson Disease: An Open-Label Pilot Study. Journal of alternative and complementary medicine (New York, N.Y.).Dec 2015;21(12):740-747.
  176. Costantini A, Fancellu R. An open-label pilot study with high-dose thiamine in Parkinson's disease. Neural Regeneration Research.12/31/accepted 2016;11(3):406-407.
  177. Walker DG et al. Gene expression changes by amyloid beta peptide-stimulated human postmortem brain microglia identify activation of multiple inflammatory processes. J Leukoc Biol. 2006 Mar;79(3):596-610. Epub 2005 Dec 19.
  178. Guillot X et al. Vitamin D and inflammation. Joint Bone Spine. 2010 Dec;77(6):552-7. Epub 2010 Nov 9.
  179. Knekt P et al. Serum vitamin D and the risk of Parkinson disease. Arch Neurol. 2010 Jul;67(7):808-11.
  180. Evatt ML et al. Prevalence of vitamin d insufficiency in patients with Parkinson disease and Alzheimer disease. Arch Neurol. 2008 Oct;65(10):1348-52.
  181. Virmani A, Gaetani F, Imam S, Binienda Z, Ali S. The protective role of L-carnitine against neurotoxicity evoked by drug of abuse, methamphetamine, could be related to mitochondrial dysfunction. Ann N Y Acad Sci. Jun 2002;965:225-232.
  182. Bodis-Wollner I, Chung E, Ghilardi MF, et al. Acetyl-levo-carnitine protects against MPTP-induced parkinsonism in primates. J Neural Transm Park Dis Dement Sect. 1991;3(1):63-72.
  183. Virmani  A, Gaetani F, Binienda Z. Effects of metabolic modifiers such as carnitines, coenzyme Q10, and PUFAs against different forms of neurotoxic insults: metabolic inhibitors, MPTP, and methamphetamine. Ann N Y Acad Sci. Aug 2005;1053:183-191.
  184. Wang C, Sadovova N, Ali HK, et al. L-carnitine protects neurons from 1-methyl-4-phenylpyridinium-induced neuronal apoptosis in rat forebrain culture. Neuroscience. Jan 5 2007;144(1):46-55.
  185. Zhang H, Jia H, Liu J, et al. Combined R-alpha-lipoic acid and acetyl-L-carnitine exerts efficient preventative effects in a cellular model of Parkinson's disease. J Cell Mol Med. 2010 Jan;14(1-2):215-25.
  186. Mandel SA, Amit T, Weinreb O, Reznichenko L, Youdim MB. Simultaneous manipulation of multiple brain targets by green tea catechins: a potential neuroprotective strategy for Alzheimer and Parkinson diseases. CNS Neurosci Ther. Winter 2008;14(4):352-365.
  187. Levites Y, Weinreb O, Maor G, Youdim MB, Mandel S. Green tea polyphenol (-)-epigallocatechin-3-gallate prevents N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced dopaminergic neurodegeneration. J Neurochem. Sep 2001;78(5):1073-1082.
  188. Pan T, Jankovic J, Le W. Potential therapeutic properties of green tea polyphenols in Parkinson's disease. Drugs Aging. 2003;20(10):711-721.
  189. Levites Y, Amit T, Youdim MB, Mandel S. Involvement of protein kinase C activation and cell survival/ cell cycle genes in green tea polyphenol (-)-epigallocatechin 3-gallate neuroprotective action. J Biol Chem. Aug 23 2002;277(34):30574-30580.
  190. Choi JY, Park CS, Kim DJ, et al. Prevention of nitric oxide-mediated 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced Parkinson's disease in mice by tea phenolic epigallocatechin 3-gallate. Neurotoxicology. Sep 2002;23(3):367-374.
  191. Nie G, Cao Y, Zhao B. Protective effects of green tea polyphenols and their major component, (-)-epigallocatechin-3-gallate (EGCG), on 6-hydroxydopamine-induced apoptosis in PC12 cells. Redox Rep. 2002;7(3):171-177.
  192. Mandel S, Maor G, Youdim MB. Iron and alpha-synuclein in the substantia nigra of MPTP-treated mice: effect of neuroprotective drugs R-apomorphine and green tea polyphenol (-)-epigallocatechin-3-gallate. J Mol Neurosci. 2004;24(3):401-416.
  193. Guo S, Bezard E, Zhao B. Protective effect of green tea polyphenols on the SH-SY5Y cells against 6-OHDA induced apoptosis through ROS-NO pathway. Free Radic Biol Med. Sep 1 2005;39(5):682-695.
  194. Guo S, Yan J, Yang T, Yang X, Bezard E, Zhao B. Protective effects of green tea polyphenols in the 6-OHDA rat model of Parkinson's disease through inhibition of ROS-NO pathway. Biol Psychiatry. Dec 15 2007;62(12):1353-1362.
  195. Li R et al. (-)-Epigallocatechin gallate inhibits lipopolysaccharide-induced microglial activation and protects against inflammation-mediated dopaminergic neuronal injury. J Neurosci Res. 2004 Dec 1;78(5):723-31.
  196. Cho HS, Kim S, Lee SY, Park JA, Kim SJ, Chun HS. Protective effect of the green tea component, L-theanine on environmental toxins-induced neuronal cell death. Neurotoxicology. Jul 2008;29(4):656-662.
  197. Mandel SA, Amit T, Kalfon L, Reznichenko L, Youdim MB. Targeting multiple neurodegenerative diseases etiologies with multimodal-acting green tea catechins. J Nutr. Aug 2008;138(8):1578S-1583S.
  198. Li R, Peng N, Du F, Li XP, Le WD. Epigallocatechin gallate protects dopaminergic neurons against 1-methyl-4- phenyl-1,2,3,6-tetrahydropyridine-induced neurotoxicity by inhibiting microglial cell activation. Nan Fang Yi Ke Da Xue Xue Bao. Apr 2006;26(4):376-380.
  199. Ramassamy C. Emerging role of polyphenolic compounds in the treatment of neurodegenerative diseases: a review of their intracellular targets. Eur J Pharmacol. Sep 1 2006;545(1):51-64.
  200. Avramovich-Tirosh Y, Reznichenko L, Mit T, et al. Neurorescue activity, APP regulation and amyloid-beta peptide reduction by novel multi-functional brain permeable iron- chelating- antioxidants, M-30 and green tea polyphenol, EGCG. Curr Alzheimer Res. Sep 2007;4(4):403-411.
  201. Zhao B. Natural antioxidants protect neurons in Alzheimer's disease and Parkinson's disease. Neurochem Res. Apr 2009;34(4):630-638.
  202. Penumathsa SV, Maulik N. Resveratrol: a promising agent in promoting cardioprotection against coronary heart disease. Can J Physiol Pharmacol. Apr 2009;87(4):275-286.
  203. Pallas M, Casadesus G, Smith MA, et al. Resveratrol and neurodegenerative diseases: activation of SIRT1 as the potential pathway towards neuroprotection. Curr Neurovasc Res. Feb 2009;6(1):70-81.
  204. Pallas M, Verdaguer E, Tajes M, Gutierrez-Cuesta J, Camins A. Modulation of sirtuins: new targets for antiageing. Recent Pat CNS Drug Discov. Jan 2008;3(1):61-69.
  205. Lee MK, Kang SJ, Poncz M, Song KJ, Park KS. Resveratrol protects SH-SY5Y neuroblastoma cells from apoptosis induced by dopamine. Exp Mol Med. Jun 30 2007;39(3):376-384.
  206. Bureau G, Longpre F, Martinoli MG. Resveratrol and quercetin, two natural polyphenols, reduce apoptotic neuronal cell death induced by neuroinflammation. J Neurosci Res. Feb 1 2008;86(2):403-410.
  207. Jin F, Wu Q, Lu YF, Gong QH, Shi JS. Neuroprotective effect of resveratrol on 6-OHDA-induced Parkinson's disease in rats. Eur J Pharmacol. Dec 14 2008;600(1-3):78-82.
  208. Lieu CA et al. A water extract of Mucuna pruriens provides long-term amelioration of parkinsonism with reduced risk for dyskinesias. Parkinsonism Relat Disord. 2010 Aug;16(7):458-65. Epub 2010 May 31.
  209. Kasture S et al. Assessment of symptomatic and neuroprotective efficacy of Mucuna pruriens seed extract in rodent model of Parkinson's disease. Neurotox Res. 2009 Feb;15(2):111-22. Epub 2009 Feb 20.
  210. Katzenschlager R et al. Mucuna pruriens in Parkinson's disease: a double blind clinical and pharmacological study. J Neurol Neurosurg Psychiatry. 2004 Dec;75(12):1672-7.
  211. FAO. Food and Agriculture Organization of the United States. FAO Corporate Document Repository. Chapter 1: Cereals: Rationale for Fermentation. http://www.fao.org/docrep/x2184e/x2184e03.htm. Copyright 2016. Accessed 6/28/2016.
  212. Rasane P, Jha A, Sabikhi L, Kumar A, Unnikrishnan VS. Nutritional advantages of oats and opportunities for its processing as value added foods - a review. Journal of Food Science and Technology. 2015;52(2):662-675.
  213. Chen CY, Milbury PE, Collins FW, Blumberg JB. Avenanthramides are bioavailable and have antioxidant activity in humans after acute consumption of an enriched mixture from oats. The Journal of nutrition. Jun 2007;137(6):1375-1382.
  214. Ahmad M, Dar Z, Habib M. A review on Oat (Avena sativa L.) as a dual-purpose crop. Scientific Research and Essays. 2014;9(4):52-59.
  215. Peterson DM, Hahn MJ, Emmons CL. Oat avenanthramides exhibit antioxidant activities in vitro. Food Chemistry. 2002;79(4):473-478.
  216. Fagervall I, Ross SB. A and B forms of monoamine oxidase within the monoaminergic neurons of the rat brain. Journal of neurochemistry. Aug 1986;47(2):569-576.
  217. Kumar MJ, Andersen JK. Perspectives on MAO-B in aging and neurological disease: where do we go from here? Molecular neurobiology. Aug 2004;30(1):77-89.
  218. Shih JC, Chen K, Ridd MJ. Monoamine oxidase: from genes to behavior. Annual review of neuroscience. 1999;22:197-217.
  219. Wolters E, Lees AJ, Volkmann J, van Laar T, Hovestadt A. Managing Parkinson's disease with continuous dopaminergic stimulation. CNS spectrums. Apr 2008;13(4 Suppl 7):1-14; quiz 15-16.
  220. Goldenberg MM. Medical management of Parkinson's disease. P & T : a peer-reviewed journal for formulary management. Oct 2008;33(10):590-606.
  221. Huot P, Fox SH, Brotchie JM. Dopamine Reuptake Inhibitors in Parkinson's Disease: A Review of Nonhuman Primate Studies and Clinical Trials. J Pharmacol Exp Ther. Jun 2016;357(3):562-569.
  222. Riederer P, Laux G. MAO-inhibitors in Parkinson's Disease. Experimental Neurobiology. 2011;20(1):1-17.
  223. Follmer C. Monoamine oxidase and alpha-synuclein as targets in Parkinson's disease therapy. Expert review of neurotherapeutics. Jun 2014;14(6):703-716.
  224. Gold Standard. Drug Monograph. Selegiline. www.clinicalkey.com. Last updated 12/2/2015. Accessed 6/28/2016.
  225. Jenner P. Preclinical evidence for neuroprotection with monoamine oxidase-B inhibitors in Parkinson's disease. Neurology. Oct 12 2004;63(7 Suppl 2):S13-22.
  226. Wong RH, Howe PR, Bryan J, Coates AM, Buckley JD, Berry NM. Chronic effects of a wild green oat extract supplementation on cognitive performance in older adults: a randomised, double-blind, placebo-controlled, crossover trial. Nutrients. May 2012;4(5):331-342.
  227. Moccetti T, Wullschleger C, Schmidt A, Aydogan C, Kreuter M. Bioactivity-based development of a wild green oat (Avena sativa L.) extract in support of mental health disorders. Zeitschrift für Phytotherapie. 2006;27(S 1):P24.
  228. Dimpfel W, Storni C, Verbruggen M. Ingested oat herb extract (Avena sativa) changes EEG spectral frequencies in healthy subjects. Journal of alternative and complementary medicine (New York, N.Y.). May 2011;17(5):427-434.
  229. Berry NM, Robinson MJ, Bryan J, Buckley JD, Murphy KJ, Howe PR. Acute effects of an Avena sativa herb extract on responses to the Stroop Color-Word test. Journal of alternative and complementary medicine. Jul 2011;17(7):635-637.
  230. Kennedy DO, Jackson PA, Forster J, Khan J, Grothe T, Perrinjaquet-Moccetti T, Haskell-Ramsay CF. Acute effects of a wild green-oat (Avena sativa) extract on cognitive function in middle-aged adults: A double-blind, placebo-controlled, within-subjects trial. Nutritional neuroscience. Nov 30 2015.
  231. Wong RH, Howe PR, Coates AM, Buckley JD, Berry NM. Chronic consumption of a wild green oat extract (Neuravena) improves brachial flow-mediated dilatation and cerebrovascular responsiveness in older adults. Journal of hypertension. Jan 2013;31(1):192-200.
  232. Rucker R, Chowanadisai W, Nakano M. Potential physiological importance of pyrroloquinoline quinone. Alternative medicine review: a journal of clinical therapeutic.Sep 2009;14(3):268-277.
  233. Harris CB, Chowanadisai W, Mishchuk DO, Satre MA, Slupsky CM, Rucker RB. Dietary pyrroloquinoline quinone (PQQ) alters indicators of inflammation and mitochondrial-related metabolism in human subjects. The Journal of nutritional biochemistry.Dec 2013;24(12):2076-2084.
  234. Misra HS, Rajpurohit YS, Khairnar NP. Pyrroloquinoline-quinone and its versatile roles in biological processes. Journal of biosciences.Jun 2012;37(2):313-325.
  235. Wu JZ, Huang JH, Khanabdali R, Kalionis B, Xia SJ, Cai WJ. Pyrroloquinoline quinone enhances the resistance to oxidative stress and extends lifespan upon DAF-16 and SKN-1 activities in C. elegans. Exp Gerontol.Jul 2016;80:43-50.
  236. Itoh Y, Hine K, Miura H, Uetake T, Nakano M, Takemura N, Sakatani K. Effect of the Antioxidant Supplement Pyrroloquinoline Quinone Disodium Salt (BioPQQ) on Cognitive Functions. Advances in experimental medicine and biology.2016;876:319-325.
  237. Zhang Q, Ding M, Gao XR, Ding F. Pyrroloquinoline quinone rescues hippocampal neurons from glutamate-induced cell death through activation of Nrf2 and up-regulation of antioxidant genes. Genetics and molecular research: GMR.2012;11(3):2652-2664.
  238. 239. Blesa J, Trigo-Damas I, Quiroga-Varela A, Jackson-Lewis VR. Oxidative stress and Parkinson's disease. Frontiers in neuroanatomy.2015;9:91.
  239. Qin J, Wu M, Yu S, et al. Pyrroloquinoline quinone-conferred neuroprotection in rotenone models of Parkinson's disease. Toxicology letters.Nov 4 2015;238(3):70-82.
  240. Nakano M, Murayama Y, Hu L, Ikemoto K, Uetake T, Sakatani K. Effects of Antioxidant Supplements (BioPQQ) on Cerebral Blood Flow and Oxygen Metabolism in the Prefrontal Cortex. Advances in experimental medicine and biology.2016;923:215-222.
  241. Ohwada K, Takeda H, Yamazaki M, et al. Pyrroloquinoline Quinone (PQQ) Prevents Cognitive Deficit Caused by Oxidative Stress in Rats. Journal of clinical biochemistry and nutrition.Jan 2008;42:29-34.
  242. Yang C, Yu L, Kong L, et al. Pyrroloquinoline quinone (PQQ) inhibits lipopolysaccharide induced inflammation in part via downregulated NF-kappaB and p38/JNK activation in microglial and attenuates microglia activation in lipopolysaccharide treatment mice. PloS one. 2014;9(10):e109502.
  243. Guan S, Xu J, Guo Y, Ge D, Liu T, Ma X, Cui Z. Pyrroloquinoline quinone against glutamate-induced neurotoxicity in cultured neural stem and progenitor cells. International journal of developmental neuroscience: the official journal of the International Society for Developmental Neuroscience.May 2015;42:37-45.
  244. Zhang Q, Zhang J, Jiang C, Qin J, Ke K, Ding F. Involvement of ERK1/2 pathway in neuroprotective effects of pyrroloquinoline quinine against rotenone-induced SH-SY5Y cell injury. Neuroscience.Jun 13 2014;270:183-191.
  245. Kim J, Harada R, Kobayashi M, Kobayashi N, Sode K. The inhibitory effect of pyrroloquinoline quinone on the amyloid formation and cytotoxicity of truncated alpha-synuclein. Molecular neurodegeneration.2010;5:20.
  246. Kim J, Kobayashi M, Fukuda M, et al. Pyrroloquinoline quinone inhibits the fibrillation of amyloid proteins. Prion.Jan-Mar 2010;4(1):26-31.
  247. Zhang JJ, Zhang RF, Meng XK. Protective effect of pyrroloquinoline quinone against Abeta-induced neurotoxicity in human neuroblastoma SH-SY5Y cells. Neuroscience letters.Oct 30 2009;464(3):165-169.
  248. Kobayashi M, Kim J, Kobayashi N, Han S, Nakamura C, Ikebukuro K, Sode K. Pyrroloquinoline quinone (PQQ) prevents fibril formation of alpha-synuclein. Biochemical and biophysical research communications.Oct 27 2006;349(3):1139-1144.
  249. Chen J, Tang XQ, Zhi JL, et al. Curcumin protects PC12 cells against 1-methyl-4-phenylpyridinium ion-induced apoptosis by bcl-2-mitochondria-ROS-iNOS pathway. Apoptosis. Jun 2006;11(6):943-953.
  250. Jagatha B, Mythri RB, Vali S, Bharath MM. Curcumin treatment alleviates the effects of glutathione depletion in vitro and in vivo: therapeutic implications for Parkinson's disease explained via in silico studies. Free Radic Biol Med. Mar 1 2008;44(5):907-917.
  251. Mythri RB, Jagatha B, Pradhan N, Andersen J, Bharath MM. Mitochondrial complex I inhibition in Parkinson's disease: how can curcumin protect mitochondria? Antioxid Redox Signal. Mar 2007;9(3):399-408.
  252. Pandey N, Strider J, Nolan WC, Yan SX, Galvin JE. Curcumin inhibits aggregation of alpha-synuclein. Acta Neuropathol. Apr 2008;115(4):479-489.
  253. Rajeswari A, Sabesan M. Inhibition of monoamine oxidase-B by the polyphenolic compound, curcumin and its metabolite tetrahydrocurcumin, in a model of Parkinson's disease induced by MPTP neurodegeneration in mice. Inflammopharmacology. Apr 2008;16(2):96-99.
  254. Sethi P, Jyoti A, Hussain E, Sharma D. Curcumin attenuates aluminium-induced functional neurotoxicity in rats. Pharmacol Biochem Behav. Apr 16 2009.
  255. Yang S, Zhang D, Yang Z, et al. Curcumin protects dopaminergic neuron against LPS induced neurotoxicity in primary rat neuron/glia culture. Neurochem Res. Oct 2008;33(10):2044-2053.
  256. Zbarsky V, Datla KP, Parkar S, Rai DK, Aruoma OI, Dexter DT. Neuroprotective properties of the natural phenolic antioxidants curcumin and naringenin but not quercetin and fisetin in a 6-OHDA model of Parkinson's disease. Free Radic Res. Oct 2005;39(10):1119-1125.
  257. Capitelli C, Sereniki A, Lima MM, Reksidler AB, Tufik S, Vital MA. Melatonin attenuates tyrosine hydroxylase loss and hypolocomotion in MPTP-lesioned rats. Eur J Pharmacol. Oct 10 2008;594(1-3):101-108.
  258. Dowling GA, Mastick J, Colling E, Carter JH, Singer CM, Aminoff MJ. Melatonin for sleep disturbances in Parkinson's disease. Sleep Med. Sep 2005;6(5):459-466.
  259. Klongpanichapak S, Phansuwan-Pujito P, Ebadi M, Govitrapong P. Melatonin inhibits amphetamine-induced increase in alpha-synuclein and decrease in phosphorylated tyrosine hydroxylase in SK-N-SH cells. Neurosci Lett. May 16 2008;436(3):309-313.
  260. Lin CH, Huang JY, Ching CH, Chuang JI. Melatonin reduces the neuronal loss, downregulation of dopamine transporter, and upregulation of D2 receptor in rotenone-induced parkinsonian rats. J Pineal Res. Mar 2008;44(2):205-213.
  261. Ma J, Shaw VE, Mitrofanis J. Does melatonin help save dopaminergic cells in MPTP-treated mice? Parkinsonism Relat Disord. May 2009;15(4):307-314.
  262. Medeiros CA, Carvalhedo de Bruin PF, Lopes LA, Magalhaes MC, de Lourdes Seabra M, de Bruin VM. Effect of exogenous melatonin on sleep and motor dysfunction in Parkinson's disease. A randomized, double blind, placebo-controlled study. J Neurol. Apr 2007;254(4):459-464.
  263. Paus S, Schmitz-Hubsch T, Wullner U, Vogel A, Klockgether T, Abele M. Bright light therapy in Parkinson's disease: a pilot study. Mov Disord. Jul 30 2007;22(10):1495-1498.
  264. Saravanan KS, Sindhu KM, Mohanakumar KP. Melatonin protects against rotenone-induced oxidative stress in a hemiparkinsonian rat model. J Pineal Res. Apr 2007;42(3):247-253.
  265. Willis GL. Parkinson's disease as a neuroendocrine disorder of circadian function: dopamine-melatonin imbalance and the visual system in the genesis and progression of the degenerative process. Rev Neurosci. 2008;19(4-5):245-316.
  266. Willis GL, Turner EJ. Primary and secondary features of Parkinson's disease improve with strategic exposure to bright light: a case series study. Chronobiol Int. 2007;24(3):521-537.
  267. Clark J et al. Oral N-acetyl-cysteine attenuates loss of dopaminergic terminals in alpha-synuclein overexpressing mice. PLoS One. 2010 Aug 23;5(8):e12333.
  268. Jana S et al. Mitochondrial dysfunction mediated by quinone oxidation products of dopamine: Implications in dopamine cytotoxicity and pathogenesis of Parkinson's disease. Biochim Biophys Acta. 2011 Jun;1812(6):663-73. Epub 2011 Mar 4.
  269. Ghibu E et al. Antioxidant properties of an endogenous thiol: Alpha-lipoic acid, useful in the prevention of cardiovascular diseases. J Cardiovasc Pharmacol. 2009 Nov;54(5):391-8.
  270. De Araujo DP et al. The contributions of antioxidant activity of lipoic acid in reducing neurogenerative progression of Parkinson's disease: a review. Int J Neurosci. 2011 Feb;121(2):51-7. Epub 2010 Dec 2.
  271. Cassani E et al. Use of probiotics for the treatment of constipation in Parkinson's disease patients. Minerva Gastroenterol Dietol. 2011 Jun;57(2):117-21.
  272. Park YK, Chung YS, Kim YS, Kwon OY, Joh TH. Inhibition of gene expression and production of iNOS and TNF-alpha in LPS-stimulated microglia by methanol extract of Phellodendri cortex. International immunopharmacology. Jul 2007;7(7):955-62. doi:10.1016/j.intimp.2006.03.018
  273. Zarmouh NO, Messeha SS, Elshami FM, Soliman KF. Natural Products Screening for the Identification of Selective Monoamine Oxidase-B Inhibitors. European J Med Plants. May 2016;15(1)doi:10.9734/ejmp/2016/26453
  274. Mazzio E, Deiab S, Park K, Soliman KF. High throughput screening to identify natural human monoamine oxidase B inhibitors. Phytother Res. Jun 2013;27(6):818-28. doi:10.1002/ptr.4795
  275. Marsh EN, Meléndez GD. Adenosylcobalamin enzymes: theory and experiment begin to converge. Biochim Biophys Acta. Nov 2012;1824(11):1154-64. doi:10.1016/j.bbapap.2012.03.012
  276. Paul C, Brady DM. Comparative Bioavailability and Utilization of Particular Forms of B(12) Supplements With Potential to Mitigate B(12)-related Genetic Polymorphisms. Integrative medicine (Encinitas, Calif). Feb 2017;16(1):42-49.
  277. Hannibal L, DiBello PM, Yu M, et al. The MMACHC proteome: hallmarks of functional cobalamin deficiency in humans. Molecular genetics and metabolism. Jul 2011;103(3):226-39. doi:10.1016/j.ymgme.2011.03.008
  278. Schaffner A, Li X, Gomez-Llorente Y, et al. Vitamin B(12) modulates Parkinson's disease LRRK2 kinase activity through allosteric regulation and confers neuroprotection. Cell research. Apr 2019;29(4):313-329. doi:10.1038/s41422-019-0153-8
  279. Hur EM, Lee BD. LRRK2 at the Crossroad of Aging and Parkinson's Disease. Genes. Mar 29 2021;12(4)doi:10.3390/genes12040505
  280. Marchand A, Drouyer M, Sarchione A, Chartier-Harlin MC, Taymans JM. LRRK2 Phosphorylation, More Than an Epiphenomenon. Front Neurosci. 2020;14:527. doi:10.3389/fnins.2020.00527
  281. Schneider SA, Hizli B, Alcalay RN. Emerging Targeted Therapeutics for Genetic Subtypes of Parkinsonism. Neurotherapeutics : the journal of the American Society for Experimental NeuroTherapeutics. Oct 2020;17(4):1378-1392. doi:10.1007/s13311-020-00920-8
  282. Koushki K, Shahbaz SK, Mashayekhi K, et al. Anti-inflammatory Action of Statins in Cardiovascular Disease: the Role of Inflammasome and Toll-Like Receptor Pathways. Clin Rev Allergy Immunol. Apr 2021;60(2):175-199. doi:10.1007/s12016-020-08791-9
  283. Kosowski M, Smolarczyk-Kosowska J, Hachula M, et al. The Effects of Statins on Neurotransmission and Their Neuroprotective Role in Neurological and Psychiatric Disorders. Molecules (Basel, Switzerland). May 11 2021;26(10)doi:10.3390/molecules26102838
  284. Yan J, Qiao L, Wu J, Fan H, Sun J, Zhang Y. Simvastatin Protects Dopaminergic Neurons Against MPP+-Induced Oxidative Stress and Regulates the Endogenous Anti-Oxidant System Through ERK. Cellular physiology and biochemistry : international journal of experimental cellular physiology, biochemistry, and pharmacology. 2018;51(4):1957-1968. doi:10.1159/000495720
  285. Bai S, Song Y, Huang X, et al. Statin Use and the Risk of Parkinson's Disease: An Updated Meta-Analysis. PLoS One. 2016;11(3):e0152564. doi:10.1371/journal.pone.0152564
  286. Yan J, Qiao L, Tian J, et al. Effect of statins on Parkinson's disease: A systematic review and meta-analysis. Medicine. Mar 2019;98(12):e14852. doi:10.1097/MD.0000000000014852
  287. Liu G, Sterling NW, Kong L, et al. Statins may facilitate Parkinson's disease: Insight gained from a large, national claims database. Mov Disord. Jun 2017;32(6):913-917. doi:10.1002/mds.27006
  288. Carroll CB, Webb D, Stevens KN, et al. Simvastatin as a neuroprotective treatment for Parkinson's disease (PD STAT): protocol for a double-blind, randomised, placebo-controlled futility study. BMJ open. Oct 7 2019;9(10):e029740. doi:10.1136/bmjopen-2019-029740
  289. Manzar H, Abdulhussein D, Yap TE, Cordeiro MF. Cellular Consequences of Coenzyme Q10 Deficiency in Neurodegeneration of the Retina and Brain. International journal of molecular sciences. 2020;21(23):9299.
  290. Trist BG, Hare DJ, Double KL. Oxidative stress in the aging substantia nigra and the etiology of Parkinson's disease. Aging Cell. Dec 2019;18(6):e13031. doi:10.1111/acel.13031
  291. Park HW, Park CG, Park M, et al. Intrastriatal administration of coenzyme Q10 enhances neuroprotection in a Parkinson's disease rat model. Sci Rep. Jun 12 2020;10(1):9572. doi:10.1038/s41598-020-66493-w
  292. Charvin D, Medori R, Hauser RA, Rascol O. Therapeutic strategies for Parkinson disease: beyond dopaminergic drugs. Nat Rev Drug Discov. Nov 2018;17(11):804-822. doi:10.1038/nrd.2018.136
  293. Parkinson Study Group QEI, Beal MF, Oakes D, et al. A randomized clinical trial of high-dosage coenzyme Q10 in early Parkinson disease: no evidence of benefit. JAMA Neurol. May 2014;71(5):543-52. doi:10.1001/jamaneurol.2014.131
  294. Negida A, Menshawy A, El Ashal G, et al. Coenzyme Q10 for Patients with Parkinson's Disease: A Systematic Review and Meta-Analysis. CNS & neurological disorders drug targets. 2016;15(1):45-53. doi:10.2174/1871527314666150821103306
  295. Zhu ZG, Sun MX, Zhang WL, Wang WW, Jin YM, Xie CL. The efficacy and safety of coenzyme Q10 in Parkinson's disease: a meta-analysis of randomized controlled trials. Neurol Sci. Feb 2017;38(2):215-224. doi:10.1007/s10072-016-2757-9
  296. Mantle D, Heaton RA, Hargreaves IP. Coenzyme Q10 and Immune Function: An Overview. Antioxidants (Basel, Switzerland). May 11 2021;10(5)doi:10.3390/antiox10050759
  297. Pradhan N, Singh C, Singh A. Coenzyme Q10 a mitochondrial restorer for various brain disorders. Naunyn-Schmiedeberg's archives of pharmacology. Nov 2021;394(11):2197-2222. doi:10.1007/s00210-021-02161-8
  298. Rabanal-Ruiz Y, Llanos-González E, Alcain FJ. The Use of Coenzyme Q10 in Cardiovascular Diseases. Antioxidants (Basel, Switzerland). May 10 2021;10(5)doi:10.3390/antiox10050755
  299. Darrow DP. Focused Ultrasound for Neuromodulation. Neurotherapeutics : the journal of the American Society for Experimental NeuroTherapeutics. Jan 2019;16(1):88-99. doi:10.1007/s13311-018-00691-3
  300. Arif WM, Elsinga PH, Gasca-Salas C, et al. Focused ultrasound for opening blood-brain barrier and drug delivery monitored with positron emission tomography. Journal of controlled release : official journal of the Controlled Release Society. Aug 10 2020;324:303-316. doi:10.1016/j.jconrel.2020.05.020
  301. Martínez-Fernández R, Rodríguez-Rojas R, del Álamo M, et al. Focused ultrasound subthalamotomy in patients with asymmetric Parkinson's disease: a pilot study. The Lancet Neurology. 2018/01/01/ 2018;17(1):54-63. doi:https://doi.org/10.1016/S1474-4422(17)30403-9
  302. Martinez-Fernandez R, Manez-Miro JU, Rodriguez-Rojas R, et al. Randomized Trial of Focused Ultrasound Subthalamotomy for Parkinson's Disease. The New England journal of medicine. Dec 24 2020;383(26):2501-2513. doi:10.1056/NEJMoa2016311
  303. Moosa S, Martinez-Fernandez R, Elias WJ, Del Alamo M, Eisenberg HM, Fishman PS. The role of high-intensity focused ultrasound as a symptomatic treatment for Parkinson's disease. Mov Disord. Sep 2019;34(9):1243-1251. doi:10.1002/mds.27779
  304. Krauss JK, Lipsman N, Aziz T, et al. Technology of deep brain stimulation: current status and future directions. Nat Rev Neurol. Feb 2021;17(2):75-87. doi:10.1038/s41582-020-00426-z
  305. Chou KL, Tarsy D. Device-assisted and lesioning procedures for Parkinson disease. UpToDate. Updated 09/02/2021. Accessed 10/25/2021, https://www.uptodate.com/contents/device-assisted-and-lesioning-procedures-for-parkinson-disease?search=deep%20brain%20stimulation%20parkinsons&source=search_result&selectedTitle=1~150&usage_type=default&display_rank=1
  306. Hartmann CJ, Fliegen S, Groiss SJ, Wojtecki L, Schnitzler A. An update on best practice of deep brain stimulation in Parkinson's disease. Therapeutic advances in neurological disorders. 2019;12:1756286419838096. doi:10.1177/1756286419838096
  307. Cagnan H, Denison T, McIntyre C, Brown P. Emerging technologies for improved deep brain stimulation. Nat Biotechnol. Sep 2019;37(9):1024-1033. doi:10.1038/s41587-019-0244-6
  308. Bratsos S, Karponis D, Saleh SN. Efficacy and Safety of Deep Brain Stimulation in the Treatment of Parkinson's Disease: A Systematic Review and Meta-analysis of Randomized Controlled Trials. Cureus. Oct 22 2018;10(10):e3474. doi:10.7759/cureus.3474
  309. Muthuraman M, Koirala N, Ciolac D, et al. Deep Brain Stimulation and L-DOPA Therapy: Concepts of Action and Clinical Applications in Parkinson's Disease. Frontiers in neurology. 2018;9:711. doi:10.3389/fneur.2018.00711
  310. Hacker ML, Turchan M, Heusinkveld LE, et al. Deep brain stimulation in early-stage Parkinson disease: Five-year outcomes. Neurology. Jul 28 2020;95(4):e393-e401. doi:10.1212/WNL.0000000000009946
  311. Mansouri A, Taslimi S, Badhiwala JH, et al. Deep brain stimulation for Parkinson's disease: meta-analysis of results of randomized trials at varying lengths of follow-up. Journal of neurosurgery. Apr 2018;128(4):1199-1213. doi:10.3171/2016.11.JNS16715
  312. Limousin P, Foltynie T. Long-term outcomes of deep brain stimulation in Parkinson disease. Nat Rev Neurol. Apr 2019;15(4):234-242. doi:10.1038/s41582-019-0145-9
  313. Aryal S, Skinner T, Bridges B, Weber JT. The Pathology of Parkinson's Disease and Potential Benefit of Dietary Polyphenols. Molecules. Sep 24 2020;25(19)doi:10.3390/molecules25194382
  314. Strikwerda AJ, Dommershuijsen LJ, Ikram MK, Voortman T. Diet Quality and Risk of Parkinson's Disease: The Rotterdam Study. Nutrients. Nov 7 2021;13(11)doi:10.3390/nu13113970
  315. Maher P. The Potential of Flavonoids for the Treatment of Neurodegenerative Diseases. International journal of molecular sciences. 2019;20(12):3056. doi:10.3390/ijms20123056
  316. Zhang X, Du L, Zhang W, Yang Y, Zhou Q, Du G. Therapeutic effects of baicalein on rotenone-induced Parkinson's disease through protecting mitochondrial function and biogenesis. Scientific reports. 2017;7(1):9968-9968. doi:10.1038/s41598-017-07442-y
  317. Luo D, Shi Y, Wang J, et al. 7,8-dihydroxyflavone protects 6-OHDA and MPTP induced dopaminergic neurons degeneration through activation of TrkB in rodents. Neurosci Lett. May 4 2016;620:43-9. doi:10.1016/j.neulet.2016.03.042
  318. Patil SP, Jain PD, Sancheti JS, Ghumatkar PJ, Tambe R, Sathaye S. Neuroprotective and neurotrophic effects of Apigenin and Luteolin in MPTP induced parkinsonism in mice. Neuropharmacology. Nov 2014;86:192-202. doi:10.1016/j.neuropharm.2014.07.012
  319. Hamsalakshmi, Alex AM, Arehally Marappa M, Joghee S, Chidambaram SB. Therapeutic benefits of flavonoids against neuroinflammation: a systematic review. Inflammopharmacol. Jan 15 2022;doi:10.1007/s10787-021-00895-8
  320. Jung UJ, Kim SR. Beneficial Effects of Flavonoids Against Parkinson's Disease. J Med Food. May 2018;21(5):421-432. doi:10.1089/jmf.2017.4078
  321. Calis Z, Mogulkoc R, Baltaci AK. The Roles of Flavonols/Flavonoids in Neurodegeneration and Neuroinflammation. Mini Rev Med Chem. 2020;20(15):1475-1488. doi:10.2174/1389557519666190617150051
  322. Zhang X, Molsberry SA, Yeh T-S, et al. Intake of Flavonoids and Flavonoid-Rich Foods, and Mortality Risk Among Individuals With Parkinson Disease: A Prospective Cohort Study. Neurology. 2021:10.1212/WNL.0000000000013275. doi:10.1212/WNL.0000000000013275
  323. Oveisgharan S, Yu L, Barnes LL, et al. Association of Statins With Cerebral Atherosclerosis and Incident Parkinsonism in Older Adults. Neurology. 2022:10.1212/WNL.0000000000200182. doi:10.1212/WNL.0000000000200182
  324. Oertel WH, Müller H-H, Unger MM, et al. Transdermal Nicotine Treatment and Progression of Early Parkinson’s Disease. NEJM Evidence. 2023;2(9):EVIDoa2200311. doi:10.1056/EVIDoa2200311. https://evidence.nejm.org/doi/abs/10.1056/EVIDoa2200311
  325. Villafane G, Thiriez C, Audureau E, et al. High-dose transdermal nicotine in Parkinson's disease patients: a randomized, open-label, blinded-endpoint evaluation phase 2 study. European journal of neurology : the official journal of the European Federation of Neurological Societies. Jan 2018;25(1):120-127. doi:10.1111/ene.13474. https://www.ncbi.nlm.nih.gov/pubmed/28960663
  326. Vieregge A, Sieberer M, Jacobs H, Hagenah JM, Vieregge P. Transdermal nicotine in PD: a randomized, double-blind, placebo-controlled study. Neurology. Sep 25 2001;57(6):1032-5. doi:10.1212/wnl.57.6.1032. https://www.ncbi.nlm.nih.gov/pubmed/11571330
  327. Ebersbach G, Stock M, Muller J, Wenning G, Wissel J, Poewe W. Worsening of motor performance in patients with Parkinson's disease following transdermal nicotine administration. Mov Disord. Nov 1999;14(6):1011-3. doi:10.1002/1531-8257(199911)14:6<1011::aid-mds1016>3.0.co;2-f. https://www.ncbi.nlm.nih.gov/pubmed/10584678