How disordered proteins spread

This news article, written for laypeople, is about how disordered proteins spread from neuron to neuron in Huntington’s Disease, and probably in other diseases as well (such as PD, AD, PSP, and CBD). What seems to be new here is a confirmation that the misfolded protein spends part of its time outside neurons as this “opens up the possibility for therapeutics.”

http://news.stanford.edu/news/2011/febr … 21811.html

Stanford Report
February 18, 2011
Stanford researchers study how disordered proteins spread from cell to cell, potentially spreading neurodegenerative disease

Misfolded proteins can get into cells and form large aggregates by recruiting normal proteins. These aggregates are associated with neurodegenerative diseases. Stanford biology Professor Ron Kopito has found that the protein linked to Huntington’s can spread from one cell to another. His research may explain how these diseases spread through our brains, an understanding that might lead to the development of drugs to target the misfolded proteins.

By Sandeep Ravindran

One bad apple is all it takes to spoil the barrel. And one misfolded protein may be all that’s necessary to corrupt other proteins, forming large aggregations linked to several incurable neurodegenerative diseases such as Huntington’s, Parkinson’s and Alzheimer’s.

Stanford biology Professor Ron Kopito has shown that the mutant, misfolded protein responsible for Huntington’s disease can move from cell to cell, recruiting normal proteins and forming aggregations in each cell it visits.

Knowing that this protein spends part of its time outside cells “opens up the possibility for therapeutics,” he said. Kopito studies how such misfolded proteins get across a cell’s membrane and into its cytoplasm, where they can interact with normal proteins. He is also investigating how these proteins move between neuronal cells.

The ability of these proteins to move from one cell to another could explain the way Huntington’s disease spreads through the brain after starting in a specific region. Similar mechanisms may be involved in the progress of Parkinson’s and Alzheimer’s through the brain.

Kopito discussed his research Friday at the annual meeting of the American Association for the Advancement of Science in Washington, D.C.

Not all bad

Not all misfolded proteins are bad. The dogma used to be that all our proteins formed neat, well-folded structures, packed together in complexes with a large number of other proteins, Kopito said. But over the past 20 years, researchers have found that as much as 30 percent of our proteins never fold into stable structures. And even ordered proteins appear to have some disordered parts.

Disordered proteins are important for normal cellular functions. Unlike regular proteins, they interact with only one partner at a time. But they are much more dynamic, capable of several quick interactions with many different proteins. This makes them ideal for a lot of the standard communication that happens within a cell for its normal functioning, Kopito said.

But if some of our proteins are always disordered, how do our cells tell which proteins need to be properly folded, and which don’t? “It’s a big mystery,” said Kopito, and one that he’s studying. This question has implications for how people develop neurodegenerative diseases, all of which appear to be age-related.

Huntington’s disease is caused by a specific mutated protein. But the body makes this mutant protein all a person’s life, so why does that person get the disease in later adulthood? Kopito said it’s because the body’s protective mechanisms stop doing their job as we get older. He said his lab hopes to determine what these mechanisms are.

A bad influence

But it’s clear what happens when these mechanisms stop working – misfolded proteins start recruiting normal versions of the same protein and form large aggregations. The presence of these aggregations in neurons has been closely linked with several neurodegenerative diseases.

Kopito found that the mutant protein associated with Huntington’s disease can leave one cell and enter another one, stirring up trouble in each new cell as it progresses down the line. The spread of the misfolded protein may explain how Huntington’s progresses through the brain.

This disease, like Parkinson’s and Alzheimer’s, starts in one area of the brain and spreads to the rest of it. This is also similar to the spread of prions, the self-replicating proteins implicated in mad cow disease and, in humans, Creutzfeldt-Jakob disease. As the misfolded protein reaches more parts of the brain, it could be responsible for the progressive worsening of these diseases.

Now that we know that these misfolded proteins spend part of their time outside of cells, traveling from one cell to another, new drugs could target them there, Kopito said. This could help prevent or at least block the progression of these diseases.

Kopito is currently working to figure out how misfolded proteins get past cell membranes into cells in the first place. It is only once in the cell’s cytoplasm that these proteins can recruit others. So these studies could help find ways to keep these mischief-makers away from the normal proteins.

He is also collaborating with biology Professor Liqun Luo to track these proteins between cells in the well-mapped fruit fly nervous system. In the future, Kopito said he hopes to link his cell biology work to disease pathology in order to understand the role misfolded proteins play in human disease.

Sandeep Ravindran is a science-writing intern at the Stanford News Service.

Davunetide Research Update (2/8/11 webinar)

I listened to Dr. Morimoto’s presentation yesterday during the CurePSP webinar. My notes are copied below. I thought the background he gave on animal studies (transgenic mice) was fascinating. Otherwise, there wasn’t anything new for me. I was surprised he didn’t mention the pilot study at all. If any of you listened in and found something interesting, please share!

You can find a video/audio recording of the presentation on the CurePSP website here:
http://curepsp.na5.acrobat.com/p92435146/

You can find a PDF of the slides on the CurePSP website here:
http://www.psp.org/file_download/a84748 … 52e9e8ccf8

Of course the slides won’t contain the Q&A.

Robin

Davunetide Research Update
Presenter: Bruce Morimoto, PhD, Vice President, Drug Development, Allon Therapeutics, Inc.
2/8/11 Webinar

[I didn’t take notes on the background he gave on Allon Therapeutics.]

Drug development is a highly regulated and lengthy process:
Preclinical: lab and animal research
Phase I: early clinical safety testing
Phase II: patient safety; early efficacy
Phase III: efficacy; safety
Ends in new drug application, FDA review, and FDA approval

Description of PSP:
* A degenerative disease involving the brain stem, basal ganglia, and cerebellum. These regions control movement.
* Clinical symptoms (movement problems, cognitive impairment) are the apparent result of the underlying tau pathology in the brain region controlling those functions.

Image from Williams and Lees; Lancet Neurology 2009;8:270-279. Location of pathology in Richardson’s Syndrome. Pathology in upper regions gives rise to cognitive impairment. Pathology in lower regions gives rise to movement problems.

In response to both disease and injury, the brain produces a protein called ADNP (activity dependent neuroprotective protein). Image on right side shows nucleotide sequence.

Davunetide has other names, including:
NAP
NAPVSIPQ Peptide
AL-108
AL-208

Davunetide protects and repairs the cell’s scaffolding (cytoskeleton/microtubules). The image on the right shows that davunetide protected a cell from zinc chloride toxin.

Microtubules form the scaffold within the cell. Microtubules are essential for the neuronal structure and function. As the cell becomes sick through neurodegenerative, the microtubules fall apart. As that happens, you lose the structure of the neuron and the function. It begins to die. Then the tau protein becomes chemically modified and starts clumping up. This gives rise to the pathology of PSP. Images from Stamelou, et al; Brain 2010:133;1578-1590.

Animal model for tau pathology:
* Created mice which progressively develop tau pathologies and spatial memory deficit. (Both a cognitive impairment as well as tau pathology.)
* Transgenic (Tg) mice given two human tau mutations. These mutations are associated with a severe FTD phenotype. (PSP is a type of FTD.)

In a study, animals were administered davunetide intra-nasally. The chemical modification associated with tau protein was analyzed. The study’s conclusion: restoration of normal tau phosphorylation levels. The level of tau went down with davunetide. This was the first hint that davunetide might affect this pathology.

Also analyzed the tangle pathology in the mice brains. The study’s conclusion: davunetide treatment reduces the number of NFTs (neurofibrillary tangles). This means that the total amount of pathology is reduced. It’s a modest improvement but is statistically significant. Shiryaev et al (2009) Neurobiology of Disease 34, 381.

While it’s important that the treatment reduces the pathology, it’s more important that the treatment improves symptoms. The tau double mutant mice, once given davunetide, showed improved learning and memory in the Morris water maze. The tau double mutant mice performed nearly as well as normal mice.

Through these studies, we see that davunetide is having an effect on:
1- the pathway that is leading to the development of the pathology
2- the level of pathology
3- behavior

Human testing of davunetide:
1) Phase 1 clinical safety studies done in (a) healthy young, middle-aged and elderly adults, and (b) Alzheimer’s Disease
2) Phase 2 clinical safety and efficacy studies done in: (a) amnestic MCI (pre-cursor to AD). After 3-month treatment, there was memory improvement. And (b) chronic cognitively-impaired schizophrenia patients. After 3-month treatment, improvement in functional capacity (everyday activities) and possible improvement in memory.

Side effects observed in previous davunetide studies:
* nasal passage complaints such as runny nose, nasal or sinus congestion, throat or sinus pain. These were potentially related to the intranasal administration.
* headache
* dizziness. Note that many PSP patients already have gait instability, episodic dizziness, and/or history of falls.
* nausea
* excessive sweating

Rationale for study of AL-108-231 in PSP:
1) In preclinical (animal) studies, davunetide reduces the tau pathology that is seen in PSP. And reducing the pathology in animals results in an improvement in behavioral outcomes.
2) In clinical studies, davunetide is: well-tolerated with modest side effects; gets into brain; improved memory in aMCI patients (pre-Alzheimer’s Disease); and improved ability to conduct daily tasks in cognitively-impaired schizophrenia patients.

Study design for AL-108-231:
* multicenter, multinational: approximately 300 PSP patients will be treated for 1 year at about 47 clinical sites in US, Canada, Australia, Germany, UK and France.
* Placebo-controlled: 1:1 ratio
* couble-blind

Key patient qualifications:
* probable or possible diagnosis of PSP with no other neurologic disease
* 41 to 85 years old
* reliable caregiver
* patient and caregiver must be fluent in local language so that interviews can be completed
* reside outside skilled nursing home or care facility at the start of the study
* ability to take 5 steps with walker or minor assistance
* parkinson medications must be stable for 90 days and other medications must be stable for 30 days

Study assessments
* Patient and caregiver interviews: safety (adverse events, con meds) and efficacy (disease severity, daily living, cognitive, mood)
* Lab tests including nasal examination, blood tests, MRI scan, lumbar puncture (optional), eye movement (optional at some sites), DNA collection (optional)

31 sites in North America: coordinating center in North America is UCSF. Dr. Adam Boxer is the study physician.

15 sites in Europe: half the sites in Germany are up and running; half the sites in the UK are up and running; the French sites should be up and running in another month or so.

1 site in Australia: Dr. David Williams in Melbourne. Open for recruitment.

For more information:
* see clinicaltrials.gov, study NCT01110720
* contact Allon Therapeutics: Sue Anne Crocker, [email protected]

Q&A: [I re-ordered these]

Question: Do you expect that davunetide will un-do damage that has already been done?

Answer: Our expectation is to prevent additional damage from occurring. We don’t expect to un-do damage that has already been done.

Question: In the 12-week pilot, there was no evidence to suggest efficacy. Does this concern you?

Answer: No. We were not expecting to see any kind of efficacy in the 12-week pilot study. It was a small study: a total of 12 patients. Not only PSP patients but also CBS patients.

Question: Will the drug help eye movements?

Answer: We don’t know. We are including this in the study since one of the distinguishing characteristics of PSP is eye movement problems. How does the eye movement dysfunction relate to some of the other impairments in PSP?

Question: What do we expect in terms of improvements?

Answer: We don’t know. Our best guess is that it’s going to take about 12 months before we see any affect from the treatment. The gains will be modest. We need a large sample size and a lengthy study period to be sure we are seeing improvements.

Question: I infer that only the Richardson’s Syndrome type of PSP patients are included. What can you say to a family dealing with another type of PSP as to the justification for this?

Answer: Inclusion criteria is really looking at the Richardson’s Syndrome type of PSP. In a clinical trial, we need to look at a similar population of patients.

Question: Is the study relevant to FTDs?

Answer: Tau pathology isn’t unique to PSP. CBS and PNFA have tau pathology. Alzheimer’s also has tau pathology.

We need to show that davunetide is effective in PSP before we can look into other conditions.

Question: Are MSA patients included in this study?

Answer: No, PSP only.

Dr. Morimoto’s Comment: Lots of questions about specific situations. Go to clinicaltrials.gov and find the site nearest you. Ask that site about your specific situation.

Dr. Morimoto’s Comment: A number of visits need to be made to the particular site. It’s up to the site to determine if you live close enough or not. Anecdotally, a number of people are travelling a long distance to participate.

[I had a few questions that didn’t get asked. They are: ]

Question: Is it reasonable to think that 47 sites can enroll 6-7 patients each to get to 300 patients? When do you anticipate the 300th patient will enroll?

Question: When might we see published data from this study?

Question: Since the Alzheimer’s market is bigger than the PSP market, why aren’t you testing this first in the AD market?

“Davunetide Research Update” (webinar, 2-8-2011)

CurePSP (psp.org) hosted a webinar yesterday to allow researchers to give an update on the experimental drug davunetide.  The key presenter was Bruce Morimoto, PhD, Vice President, Drug Development, Allon Therapeutics, Inc., the maker of davunetide.

My notes from Dr. Morimoto’s presentation and the question-and-answer session are below.

I thought the background Dr. Morimoto gave on animal studies (transgenic mice) was fascinating.  Otherwise, there wasn’t anything new for me.  I was surprised he didn’t mention the davunetide pilot study at all.  If any of you listened in and found something interesting, please share!

Editor’s Note:  Unfortunately these links are no longer working.
You can find a video/audio recording of the presentation on the CurePSP website here:
curepsp.na5.acrobat.com/p92435146/

You can find a PDF of the slides on the CurePSP website here:
www.psp.org/file_download/a8474849-0fa9-4ed9-8659-9452e9e8ccf8

Of course the slides won’t contain the Q&A.

Robin

——————————

Davunetide Research Update
Presenter:  Bruce Morimoto, PhD, Vice President, Drug Development, Allon Therapeutics, Inc.
CurePSP Webinar
February 8, 2011

[I didn’t take notes on the background he gave on Allon Therapeutics.]

Drug development is a highly regulated and lengthy process:
Preclinical:  lab and animal research
Phase I:  early clinical safety testing
Phase II:  patient safety; early efficacy
Phase III:  efficacy; safety
Ends in new drug application, FDA review, and FDA approval

Description of PSP:
* A degenerative disease involving the brain stem, basal ganglia, and cerebellum.  These regions control movement.
* Clinical symptoms (movement problems, cognitive impairment) are the apparent result of the underlying tau pathology in the brain region controlling those functions.

Image from Williams and Lees; Lancet Neurology 2009;8:270-279.  Location of pathology in Richardson’s Syndrome.  Pathology in upper regions gives rise to cognitive impairment.  Pathology in lower regions gives rise to movement problems.

In response to both disease and injury, the brain produces a protein called ADNP (activity dependent neuroprotective protein).  Image on right side shows nucleotide sequence.

Davunetide has other names, including:
NAP
NAPVSIPQ Peptide
AL-108
AL-208

Davunetide protects and repairs the cell’s scaffolding (cytoskeleton/microtubules).  The image on the right shows that davunetide protected a cell from zinc chloride toxin.

Microtubules form the scaffold within the cell.  Microtubules are essential for the neuronal structure and function.  As the cell becomes sick through neurodegenerative, the microtubules fall apart.  As that happens, you lose the structure of the neuron and the function.  It begins to die.  Then the tau protein becomes chemically modified and starts clumping up.  This gives rise to the pathology of PSP.  Images from Stamelou, et al; Brain 2010:133;1578-1590.

Animal model for tau pathology:
* Created mice which progressively develop tau pathologies and spatial memory deficit.  (Both a cognitive impairment as well as tau pathology.)
* Transgenic (Tg) mice given two human tau mutations.  These mutations are associated with a severe FTD phenotype.  (PSP is a type of FTD.)

In a study, animals were administered davunetide intra-nasally.  The chemical modification associated with tau protein was analyzed.  The study’s conclusion:  restoration of normal tau phosphorylation levels.  The level of tau went down with davunetide.  This was the first hint that davunetide might affect this pathology.

Also analyzed the tangle pathology in the mice brains.  The study’s conclusion:  davunetide treatment reduces the number of NFTs (neurofibrillary tangles).  This means that the total amount of pathology is reduced.  It’s a modest improvement but is statistically significant.  Shiryaev et al (2009) Neurobiology of Disease 34, 381.

While it’s important that the treatment reduces the pathology, it’s more important that the treatment improves symptoms.  The tau double mutant mice, once given davunetide, showed improved learning and memory in the Morris water maze.  The tau double mutant mice performed nearly as well as normal mice.

Through these studies, we see that davunetide is having an effect on:
1- the pathway that is leading to the development of the pathology
2- the level of pathology
3- behavior

Human testing of davunetide:
1) Phase 1 clinical safety studies done in (a) healthy young, middle-aged and elderly adults, and (b) Alzheimer’s Disease
2) Phase 2 clinical safety and efficacy studies done in:  (a) amnestic MCI (pre-cursor to AD).  After 3-month treatment, there was memory improvement.  And (b) chronic cognitively-impaired schizophrenia patients.  After 3-month treatment, improvement in functional capacity (everyday activities) and possible improvement in memory.

Side effects observed in previous davunetide studies:
* nasal passage complaints such as runny nose, nasal or sinus congestion, throat or sinus pain.  These were potentially related to the intranasal administration.
* headache
* dizziness.  Note that many PSP patients already have gait instability, episodic dizziness, and/or history of falls.
* nausea
* excessive sweating

Rationale for study of AL-108-231 in PSP:
1) In preclinical (animal) studies, davunetide reduces the tau pathology that is seen in PSP.  And reducing the pathology in animals results in an improvement in behavioral outcomes.
2) In clinical studies, davunetide is:  well-tolerated with modest side effects; gets into brain; improved memory in aMCI patients (pre-Alzheimer’s Disease); and improved ability to conduct daily tasks in cognitively-impaired schizophrenia patients.

Study design for AL-108-231:
* multicenter, multinational:  approximately 300 PSP patients will be treated for 1 year at about 47 clinical sites in US, Canada, Australia, Germany, UK and France.
* Placebo-controlled: 1:1 ratio
* couble-blind

Key patient qualifications:
* probable or possible diagnosis of PSP with no other neurologic disease
* 41 to 85 years old
* reliable caregiver
* patient and caregiver must be fluent in local language so that interviews can be completed
* reside outside skilled nursing home or care facility at the start of the study
* ability to take 5 steps with walker or minor assistance
* parkinson medications must be stable for 90 days and other medications must be stable for 30 days

Study assessments
* Patient and caregiver interviews:  safety (adverse events, con meds) and efficacy (disease severity, daily living, cognitive, mood)
* Lab tests including nasal examination, blood tests, MRI scan, lumbar puncture (optional), eye movement (optional at some sites), DNA collection (optional)

31 sites in North America:  coordinating center in North America is UCSF.  Dr. Adam Boxer is the study physician.

15 sites in Europe:  half the sites in Germany are up and running; half the sites in the UK are up and running; the French sites should be up and running in another month or so.

1 site in Australia:  Dr. David Williams in Melbourne.  Open for recruitment.

For more information:
* see clinicaltrials.gov, study NCT01110720
* contact Allon Therapeutics:  Sue Anne Crocker, [email protected]

Q&A:  [I re-ordered these]

Question:  Do you expect that davunetide will un-do damage that has already been done?

Answer:  Our expectation is to prevent additional damage from occurring.  We don’t expect to un-do damage that has already been done.

Question:  In the 12-week pilot, there was no evidence to suggest efficacy.  Does this concern you?

Answer:  No.  We were not expecting to see any kind of efficacy in the 12-week pilot study.  It was a small study:  a total of 12 patients.  Not only PSP patients but also CBS patients.

Question:  Will the drug help eye movements?

Answer:  We don’t know.  We are including this in the study since one of the distinguishing characteristics of PSP is eye movement problems.  How does the eye movement dysfunction relate to some of the other impairments in PSP?

Question:  What do we expect in terms of improvements?

Answer:  We don’t know.  Our best guess is that it’s going to take about 12 months before we see any affect from the treatment.  The gains will be modest.  We need a large sample size and a lengthy study period to be sure we are seeing improvements.

Question:  I infer that only the Richardson’s Syndrome type of PSP patients are included.  What can you say to a family dealing with another type of PSP as to the justification for this?

Answer:  Inclusion criteria is really looking at the Richardson’s Syndrome type of PSP.  In a clinical trial, we need to look at a similar population of patients.

Question:  Is the study relevant to FTDs?

Answer:  Tau pathology isn’t unique to PSP.  CBS and PNFA have tau pathology.  Alzheimer’s also has tau pathology.

We need to show that davunetide is effective in PSP before we can look into other conditions.

Question:  Are MSA patients included in this study?

Answer:  No, PSP only.

Dr. Morimoto’s Comment:  Lots of questions about specific situations.  Go to clinicaltrials.gov and find the site nearest you.  Ask that site about your specific situation.

Dr. Morimoto’s Comment:  A number of visits need to be made to the particular site.  It’s up to the site to determine if you live close enough or not.  Anecdotally, a number of people are travelling a long distance to participate.

[I had a few questions that didn’t get asked.  They are: ]

Question:  Is it reasonable to think that 47 sites can enroll 6-7 patients each to get to 300 patients?  When do you anticipate the 300th patient will enroll?

Question:  What’s the baseline PSP Rating Scale score for those who participated in the pilot?  What’s the baseline MMSE score for those who participated in the pilot?

Question:  I’ve heard that we might see published data from this study in 2013.  Do you think that’s reasonable?  Are we talking the beginning of 2013 or the end of 2013?

Question:  Since the Alzheimer’s market is bigger than the PSP market, why aren’t you testing this first in the AD market?

Clinical-pathological correlates in PSP (and MSA)

This is a terrific little study done comparing brain tissue of 15 PSP cases, 12 MSA cases, 8 PD cases, and 8 healthy controls, and correlating the pathology with clinical variables (age at onset, disease duration, and symptoms).

The study was done in Australia using tissue donated to the Sydney Brain Bank. I say it was a “little” study because the previous clinical-pathological correlation study in MSA and PSP had much larger numbers — 110 PSP cases and 83 MSA cases. That larger Queen Square Brain Bank (UK) study “did not consider the pathological severity of disease or assess pathological correlations to clinical features.” (This is O’Sullivan’s “Clinical outcomes” article published in the journal Brain in 2008.  We have posted about that here.)

The symptoms studied include the:

“presence or absence of…bradykinesia and rigidity, resting tremor, postural instability, response to levodopa (L-dopa) therapy, dementia as indicated from the last Clinical Dementia Rating (CDR) score, early falls (within the first 2 years of onset), supranuclear vertical gaze abnormalities (abnormal or slow vertical gaze and/or supranuclear gaze palsy), dysarthria, dysphagia, postural hypotension, autonomic and urinary dysfunction, and gait ataxia. The severity of parkinsonism was assessed using the last Hoehn and Yahr (H&Y) score prior to death.”

Here’s some key data on the clinical variables:

Age at onset
PSP: 67 years +/- 9 years

Disease duration
PSP: 7 years +/- 4 years

Phenotype (parkinsonian: parkinsonian-plus)
PSP: 5:10

% L-dopa responsive
PSP: 47%

% demented
PSP: 67%
MSA: 0%

% early falls
PSP: 93%

% gaze abnormalities
PSP: 73%

% dysarthria
PSP: 60%

% autonomic dysfunction
PSP: 47%

% gait ataxia
PSP: 40%

The researchers divide cases by phenotype — parkinsonian or parkinsonian-plus. My assumption is that, for PSP, the parkinsonian phenotype is called PSP-Parkinsonism and the parkinsonian-plus phenotype is called RS (Richardson’s Syndrome).

“As expected, response to L-dopa was associated with a parkinsonian phenotype across all groups, although resting tremor was not associated with either phenotype. The presence of supranuclear vertical gaze abnormalities and early falls was associated with a parkinsonian-plus phenotype across groups, although autonomic dysfunction and gait ataxia were not associated with either phenotype.”

I’ve copied the abstract below.

Robin

———

Movement Disorders. 2011 Jan 21.

Clinical correlates of similar pathologies in parkinsonian syndromes.

Song YJ, Huang Y, Halliday GM.
Neuroscience Research Australia and the University of New South Wales, Randwick, New South Wales, Australia.

Abstract
BACKGROUND: There have been no previous studies assessing the severity of regional atrophy, cell loss and lesion densities between the overlapping conditions of Parkinson’s disease (PD), progressive supranuclear palsy (PSP) and multiple system atrophy (MSA) and relating these pathologies to different clinical features.

METHODS: Clinical indices and basal ganglia, brainstem, and cerebellar pathology from 43 longitudinally studied cases (PD = 8, PSP = 15, MSA = 12, controls = 8) were compared. A point-counting method was used to evaluate subregional volumes, and alpha-synuclein and phospho-tau immunohistochemistry was used to assess pathological inclusions and stage disease severity. Logistic regression analyses were used to identify pathological associations with clinical features.

RESULTS: All PD, PSP, and MSA cases had severe degeneration of the substantia nigra. Clinical features correlated with tissue loss and the severity of inclusion pathologies. Levodopa responsiveness and a lack of resting tremor was associated with preservation of pallidal volume, the presence of gait ataxia was associated with atrophy of the putamen, and the parkinsonian-plus phenotype with early falls and supranuclear vertical gaze abnormalities had more substantial midbrain atrophy and greater inclusion pathology in the caudate nucleus.

DISCUSSION: This is the first study to compare the severity of regional pathologies across parkinsonian conditions. The data show that tissue loss and inclusion densities in certain regions correlate with clinical indices, with regional volume changes likely to be the best indicator of clinical progression of disease.

Copyright © 2011 Movement Disorder Society.

PubMed ID#: 21259341 (see pubmed.gov for the abstract only)

Alpha-synuclein in spinal fluid isn’t a parkinsonism biomark

Dutch researchers found that the amount of alpha-synuclein in cerebrospinal fluid (CSF) is not a tool for differentiating those who have Parkinson’s Disease from those who have an atypical parkinsonism disorder (MSA, DLB, PSP, CBD, and vascular parkinsonism). Alpha-synuclein in CSF is also not a tool for differentiating among the atypical parkinsonism disorders. The search for a biomarker continues.

Neurobiology of Aging. 2011 Jan 12. [Epub ahead of print]

CSF alpha-synuclein does not differentiate between parkinsonian disorders.

Aerts MB, Esselink RA, Abdo WF, Bloem BR, Verbeek MM.
Department of Neurology, Parkinson Center Nijmegen (ParC), and Alzheimer Centre Nijmegen, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen Medical Centre, the Netherlands.

Abstract
Differentiating between Parkinson’s disease (PD) and atypical Parkinsonism (AP) is clinically relevant but challenging.

A timely and correct diagnosis might result in better targeted treatment strategies, adequate patient counseling, and early recognition of disease-specific complications.

We aimed to investigate whether cerebrospinal fluid (CSF) concentrations of alpha-synuclein are of additional diagnostic value. We examined 142 consecutive patients with parkinsonism, mean disease duration 39.7 mo (Parkinson’s disease (PD), n = 58; MSA, n = 47; dementia with Lewy bodies (DLB), n = 3; VaP, n = 22; progressive supranuclear palsy (PSP), n = 10; CBD, n = 2).

Gold standard was the clinical diagnosis established after 2 years of clinical follow-up.

CSF concentrations of alpha-synuclein, blood pigments and the erythrocyte count were determined.

No differences between CSF alpha-synuclein concentrations of patients with PD with the reference values from our laboratory were observed.

We neither found significant differences between patients with PD and AP nor between AP subgroups. Adjustment for age, disease severity or presence of erythrocytes or blood pigments in CSF did not alter these results.

Our results imply that CSF alpha-synuclein is currently unsuitable as biomarker to differentiate between PD and AP.

Copyright 2011 Elsevier Inc. All rights reserved.

PubMed ID#: 21236518 (see pubmed.gov for abstract only)