Striatonigral Degeneration

Back

Background

Striatonigral degeneration is a sporadic, progressive neurodegenerative disorder that represents one manifestation of multiple system atrophy (MSA). Other manifestations of multiple system atrophy are Shy-Drager syndrome, in which autonomic failure predominates, and sporadic olivopontocerebellar degeneration, which is characterized primarily by cerebellar signs. While symptoms of autonomic failure and cerebellar degeneration may be present in striatonigral degeneration, the predominant finding is parkinsonism. (See Presentation and Workup.)

The group of disorders classified under multiple system atrophy belong to a broader class called the Parkinson-plus syndromes, which have in common features such as bradykinesia and rigidity seen in Parkinson disease, combined with additional components such as autonomic dysfunction, ataxia, or dementia.

In many cases, the disease process begins with 1 of the 3 presentations predominating (ie, parkinsonism, autonomic failure, cerebellar signs) and then later converges to include a combination of all 3 plus additional degeneration of the corticospinal system, including tract and motor neuron degeneration, as well as cognitive deterioration. However, in some cases, one presentation remains dominant throughout the course of the disease, or it may be that the patient dies before additional symptoms can manifest. This has been most clearly described for the cerebellar form of multiple system atrophy. (See Etiology and Pathophysiology.)

In 1933, Sherer described 2 cases that likely represented striatonigral degeneration. However, this condition was first definitively outlined in 1961 and 1964 by Adams et al. In 1969, Graham and Oppenheimer coined the term multiple system atrophy in an effort to emphasize the common features found in all the 3 manifestations.

Thirty years later, the first consensus statement on the diagnosis of multiple system atrophy, by Gilman et al, recommended that the term striatonigral degeneration be replaced with multiple system atrophy with predominantly parkinsonian features (MSA-P) and that the term sporadic olivopontocerebellar degeneration be replaced with multiple system atrophy with predominantly cerebellar features (MSA-C).[1] The term Shy-Drager syndrome was deemed unnecessary and was excluded. The consensus group described the clinical features of the disease and set the criteria for diagnosis of possible, probable, and definite multiple system atrophy. (See Presentation and Workup.)

In 2008, the second consensus statement on the diagnosis of multiple system atrophy was published. The purpose of revisiting this topic was to incorporate advances in research, such as the identification of alpha-synuclein as a key pathologic finding, and to simplify the original diagnostic criteria.[2]

Use of the new nomenclature (MSA-P and MSA-C) became common in publications subsequent to 1999; however, the term Shy-Drager syndrome is still frequently used. Many neurologists find this a useful term for the autonomic presentation, but consideration must be given to whether subtle parkinsonian or cerebellar findings are present, in which case the MSA-P or MSA-C classifications, respectively, would be more appropriate. In the event that both parkinsonian and cerebellar signs are present, the term multiple system atrophy can be used without qualification.

Etiology and Pathophysiology

MSA-P is characterized by the presence of glial cytoplasmic inclusions (GCIs) in oligodendroglial cells. These inclusions are widely distributed throughout the brains of affected individuals. Neuronal cytoplasmic inclusions and neuronal nuclear inclusions can also be found but are far less prominent relative to GCIs. It has been suggested that the involvement of both neurons and oligodendrocytes synergistically affect the neurodegenerative process in MSA.[3] The identification of these inclusion bodies was a unifying factor in the pathologic classification of the 3 entities that now fall under the category of multiple system atrophy: MSA-P, MSA-C, and, less formally, Shy-Drager syndrome.[4, 5]

Immunostaining of inclusion bodies has revealed the presence of alpha-synuclein fibrils, which further classifies this group of disorders as synucleinopathies. (However, the significance of this finding remains unclear.) Other neurodegenerative conditions that fall under this category include Parkinson disease and diffuse Lewy body disease.[6] Alpha-synuclein, in its soluble form, is found in normal brain tissue. It is the insoluble aggregate that forms the fibrils associated with synucleinopathies that appears to be pathologic.[7] A recent theory suggests that alpha-synuclein is spread from neuron to oligodendrocyte via a prion-like process. In association with neuroinflammation, this leads to a specific pattern of neurodegeneration.[8]

In addition to inclusion bodies, microscopic evaluation of tissue reveals neuronal loss and gliosis. This is manifested at a macroscopic level as atrophy, primarily of the pons and midbrain. The substantia nigra shows loss of pigmentation, while the putamen, also atrophic, may become grayish-green in color. This pattern of degeneration is consistent with clinical findings.

Epidemiology

The prevalence of multiple system atrophy (including all 3 subtypes) in the United States is difficult to establish because the disease is frequently misdiagnosed; however, the prevalence has been estimated to be 3-5 cases per 100,000 people in the general population. International data show a prevalence of 1.86-4.9 cases per 100,000 people.[9, 10]

In one Japanese study, review of the pathologic features of 102 multiple system atrophy cases revealed that MSA-C (olivopontocerebellar type MSA) was relatively more frequent in Japan than MSA-P (striatonigral degeneration type MSA), while the converse is found in western countries.[11]

Formerly, no gender predominance was observed, but information now suggests a male predominance of approximately 2:1. Some reports show a much greater disparity, with the suggestion that males who seek treatment for autonomic symptoms, such as erectile dysfunction, may be more likely to be diagnosed.

Onset occurs most often in the sixth decade. The mean age at diagnosis is 53 years, with an age range of 33-76 years.

Prognosis

Multiple system atrophy is a progressive neurodegenerative disorder without remission. The prognosis is poor, and all 3 subtypes of multiple system atrophy have a mean survival period of less than a decade from diagnosis. In a study by Schrag et al, median survival time was 8.6 years for men and 7.3 years for women.[12]

Gender, phenotype, and gait instability do not appear to be prognostic indicators of shorter survival, as has previously been proposed. However, autonomic failure continues to be a negative prognostic factor. Data from one study suggest that early development of severe generalized autonomic failure more than triples the risk of shorter survival in patients with MSA.[13]

The most marked clinical deterioration is seen in MSA-P, although the time frame for survival in this disease is similar to that of the other 2 subtypes.

Morbidity and mortality

Complications of multiple system atrophy include the following:

Patient Education

Patients with symptomatic postural hypotension should be educated with regard to the following:

History

Parkinsonism

The vast majority of patients with multiple system atrophy develop parkinsonism at some point, and the condition is often rapidly progressive.

Bradykinesia with rigidity, tremor, or postural instability is seen. Although the presentation can be asymmetrical (as is usually the case in Parkinson disease), symmetry of onset is particularly suggestive of multiple system atrophy with predominantly parkinsonian features (MSA-P). Absence of tremor is also suggestive of MSA-P (vs Parkinson disease). When present, tremor is usually irregular, postural, and may be associated with myoclonus. While resting tremor can be observed, it is uncommon.

Multiple system atrophy patients with parkinsonism may have a limited response to a trial of levodopa. However, a clear-cut response that is sustained beyond 2 years is unusual.

Dysautonomia

Some degree of autonomic failure is almost universal and may be the presenting symptom. Genitourinary complaints are common early in the disease. Symptoms include the following:

Sleep disturbances

When possible, obtain information on the patient’s history from the patient’s sleeping partner. Sleep-related symptoms include the following:

Additional findings

Additional findings in MSA-P include the following:

Other etiologies

Features that suggest an etiology other than multiple system atrophy include the following:

Physical Examination

Parkinsonism

Bradykinesia with rigidity, tremor, or postural instability occurs. Symmetrical onset is suggestive of multiple system atrophy (vs Parkinson disease). Absence of tremor is suggestive of MSA-P (vs Parkinson disease). When present, tremor is usually irregular, postural, and is associated with myoclonus. While resting tremor can be observed, it is uncommon.

Dysautonomia

Symptoms of dysautonomia include the following:

Cerebellar findings

Cerebellar findings include the following:

Cognitive impairment

Although dementia (as a predominant feature) is a criterion for exclusion of multiple system atrophy, studies suggest that some degree of cognitive impairment is common in multiple system atrophy, and particularly so in MSA-P.[16] The extent of impairment varies significantly. When present, deficits include the following:

Additional findings

Additional findings in MSA-P include the following:

Other etiologies

Features that suggest an etiology other than multiple system atrophy include the following:

Approach Considerations

No laboratory studies are diagnostic for multiple system atrophy with predominantly parkinsonian features (MSA-P). If a family history is noted, then testing for the SCA-3 mutation can identify Machado-Joseph disease.

Electromyography

Electromyography shows denervation of the external sphincter (urethral or anal); however, normal findings do not exclude the disease.

Clonidine growth hormone test

Studies show that after infusion of clonidine, serum growth hormone concentration does not subsequently rise in patients with multiple system atrophy. In contrast, the normal response, an increase in secretion, is found in Parkinson disease, pure autonomic failure, and control subjects.[18, 19]

Neuropsychiatric evaluation

Studies suggest that cognitive impairment is more common than previously thought in multiple system atrophy. Due to the nature of the deficits associated with this disease, the standard mental status examination has been found to be a poor tool for assessment. Neuropsychiatric testing is more sensitive and may be more a more helpful resource in multiple system atrophy.[16]

Sleep studies

Sleep disorders, particularly nocturnal stridor and REM sleep behavior disorder, are common in multiple system atrophy. Formal sleep studies should be considered, as research suggests that treatment can improve survival and quality of life.[20]

REM sleep behavior disorder is common in Parkinson disease and atypical parkinsonian disorders including striatonigral degeneration (MSA-P). Clonazepam is an effective treatment for many patients. Nocturnal stridor is specific to MSA and is produced by vocal cord dysfunction during sleep. CPAP is appropriate for long term treatment.[21]

Histologic findings

Findings in MSA-P include widespread glial cytoplasmic inclusions (primarily in oligodendrocytes) and, to a lesser degree, neuronal cytoplasmic inclusions and neuronal nuclear inclusions. Immunostaining of inclusion bodies reveals the presence of alpha-synuclein fibrils.[22]

Other tests

Autonomic tests for orthostatic vital signs such as the tilt-table test and urodynamic studies can be conducted. Scintigraphy with iodine-123-metaiodobenzylguanidine (MIBG) has recently shown utility in the differential diagnosis between Parkinson disease and multiple system atrophy; this method shows high sensitivity and adequate specificity in this field.[23]

A study measuring the clinical utility of skin biopsy for differentiating between Parkinson disease and multiple system atrophy concluded that detection of alpha-synuclein aggregates on cutaneous nerves in distal body sites is insufficiently sensitive; however, intraepidermal nerve fiber density (IEND) may be useful for this purpose. Further study is needed.[24]

Imaging Studies

Computed tomography (CT) scans may show cerebellar or brainstem atrophy late in the course of the disease.

A study using single-photon emission CT (SPECT) scanning revealed significantly decreased cerebellar and dorsolateral prefrontal perfusion in patients with multiple system atrophy, relative to that of control subjects.[16]

Magnetic resonance imaging (MRI) may show 1 or more of the following[25] :

Positron emission tomography (PET) scans demonstrate decreased postsynaptic D2-receptor density and impaired uptake of fluoro-L-dopa.[27] The recently approved DaTscan single-photon emission computed tomography (SPECT) imaging can help confirm the diagnosis of parkinsonism but does not distinguish idiopathic Parkinson disease from multiple system atrophy.[28]

Approach Considerations

In patients with multiple system atrophy, response to antiparkinsonian medications is suboptimal at best. However, these drugs remain the treatment of choice in the absence of better options.[29] Other medical therapies used in multiple system atrophy target associated symptoms (eg, orthostatic hypotension).[30] Surgical treatment

Currently, no surgical treatment is appropriate for multiple system atrophy. Because it can be difficult to clinically distinguish multiple system atrophy from Parkinson disease, there are cases of multiple system atrophy patients undergoing placement of deep brain stimulators. The outcomes have generally been poor, even in patients who responded well to levodopa therapy.[31, 32]

Inpatient care

Multiple system atrophy with predominantly parkinsonian features (MSA-P) is unlikely to be the primary cause of a patient’s hospitalization. Thus, the focus of care would be treatment of the diagnoses that required admission.

Outpatient care

Outpatient care includes the following:

Consultations

Consultations in multiple system atrophy can include professionals in the following specialties:

Diet

Unless there are contraindications, patients with symptomatic postural hypotension may benefit from increased salt intake.

Activity

Patients with symptomatic postural hypotension should be advised to avoid activities or environments that produce excessive vagal stimulation or vasodilation (eg, extreme heat, overeating, straining at stool) and to rise slowly and carefully from seated or recumbent positions.

Medication Summary

The drugs in the tables below are specific to the treatment of parkinsonism and postural hypotension associated with multiple system atrophy with predominantly parkinsonian features (MSA-P). As previously mentioned, in patients with multiple system atrophy, the response to antiparkinsonian medications is suboptimal at best. Because better options are not available, however, these agents remain the treatment of choice for the disease.

Adjunct medications

Anticholinergic medications, such as oxybutynin, are sometimes used for incontinence but often lead to subsequent retention. Although sildenafil has been used for treatment of erectile dysfunction, it is generally not recommended, due to its high potential to provoke or exacerbate hypotension. The use of a fiber supplement or another bowel regimen may be necessary for constipation.

A selective serotonin reuptake inhibitor (SSRI) or similar drug may be required for the treatment of depression often associated with all subtypes of multiple system atrophy. For patients who suffer from REM sleep behavioral disorder, clonazepam may be beneficial. Botulinum toxin (BOTOX®) injection to the vocal cords has been used for the treatment of stridor.

Levodopa-carbidopa (Sinemet, Parcopa)

Clinical Context:  Levodopa is a dopamine precursor used to increase central nervous system (CNS) dopamine concentration, as it is not possible for dopamine to cross the blood-brain barrier. Carbidopa is a peripheral dopa decarboxylase inhibitor that prevents premature conversion of levodopa to dopamine in the tissues prior to entering the CNS. It increases the efficiency of levodopa therapy, allows for lower dosages, and also decreases the side effects associated with peripheral conversion.

Standard release forms of levodopa-carbidopa are available in 25/100-, 10/100-, and 25/250-mg tablets. Controlled-release preparations are available in 50/200 mg and 25/100 mg.

Pramipexole (Mirapex)

Clinical Context:  Pramipexole is a nonergot dopamine agonist that is used with or without concomitant levodopa therapy. It binds D2 and D3 dopamine receptors. Due to pramipexole's high specificity for D3 receptors (relative to other dopamine agonists), it may cause less orthostatic hypotension. It has no significant effect on other adrenergic or serotonergic receptors. The drug's absolute bioavailability is greater than 90%. Its peak serum concentration is reached in approximately 2 hours and its half-life is approximately 8 hours.

There are no known metabolites; roughly 90% of this drug is renally excreted in its unchanged form. Tablets are available in 0.125-, 0.25-, 0.5-, 0.75-, 1-, and 1.5-mg forms.

Ropinirole (Requip)

Clinical Context:  Ropinirole is a nonergot dopamine agonist that is used with or without concomitant levodopa therapy. It binds to D2 and D3 receptors but has a greater affinity for D3. Ropinirole's bioavailability is 55%, its peak plasma concentration is reached in 1-2 hours, and its half-life is approximately 6 hours. Ropinirole is extensively metabolized by the liver via P450 CYP1A2. Less than 10% of the drug is renally excreted; no dosage change is required in mild to moderate renal insufficiency. If ropinirole is used as adjunct therapy, it may be possible to titrate levodopa dosage slowly downward.

Class Summary

Dopaminergic drugs can exacerbate orthostatic hypotension. They must be initiated at low doses and cautiously titrated up.

Fludrocortisone

Clinical Context:  Fludrocortisone is a synthetic steroid with predominantly mineralocorticoid activity. It acts on renal distal tubules to enhance the reabsorption of sodium and increase the urinary excretion of potassium. The net effect is an increase in plasma volume and an elevation of blood pressure. The drug's metabolism is primarily hepatic.

Class Summary

These are used to treat orthostatic hypotension that is refractory to nonpharmacologic recommendations.

Midodrine

Clinical Context:  Midodrine is a selective alpha1-adrenergic agonist used for the treatment of hypotension.

Class Summary

These drugs are used to treat orthostatic hypotension that is refractory to nonpharmacologic recommendations.

Clonazepam (Klonopin)

Clinical Context:  Clonazepam is a benzodiazepine which binds to gamma aminobutyric acid (GABA)-A receptors in the CNS. It may help regulate sleep disorders.

Class Summary

These drugs are used for REM sleep behavior disorder (RBD).

Author

Ahmad El Kouzi, MD, Resident Physician, Department of Neurology, Southern Illinois University School of Medicine

Disclosure: Nothing to disclose.

Coauthor(s)

Paula K Rauschkolb, DO, Assistant Professor of Neurology and Medicine, Geisel School of Medicine at Dartmouth; Consulting Staff Physician, Department of Neurology, Department of Medicine, Section of Hematology/Oncology, Dartmouth-Hitchcock Medical Center

Disclosure: Nothing to disclose.

Stephen A Berman, MD, PhD, MBA, Professor of Neurology, University of Central Florida College of Medicine

Disclosure: Nothing to disclose.

Chief Editor

Selim R Benbadis, MD, Professor, Director of Comprehensive Epilepsy Program, Departments of Neurology and Neurosurgery, Tampa General Hospital, University of South Florida Morsani College of Medicine

Disclosure: Serve(d) as a director, officer, partner, employee, advisor, consultant or trustee for: Ceribell, Eisai, Greenwich, Growhealthy, LivaNova, Neuropace, SK biopharmaceuticals, Sunovion<br/>Serve(d) as a speaker or a member of a speakers bureau for: Eisai, Greenwich, LivaNova, Sunovion<br/>Received research grant from: Cavion, LivaNova, Greenwich, Sunovion, SK biopharmaceuticals, Takeda, UCB.

Acknowledgements

Maritza Arroyo-Muñiz, MD Associate Program Director, Professor of Neurology, Department of Neurology, University of Puerto Rico

Maritza Arroyo-Muñiz, MD is a member of the following medical societies: American Academy of Neurology, National Stroke Association

Disclosure: Nothing to disclose.

Syed T Arshad, MD Staff Physician, Department of Neurology, Dartmouth Hitchcock Medical Center

Syed T Arshad, MD is a member of the following medical societies: American Academy of Family Physicians, American Medical Association

Disclosure: Nothing to disclose.

Arif I Dalvi, MD Director, Movement Disorders Center, NorthShore University HealthSystem, Clinical Associate Professor of Neurology, University of Chicago Pritzker Medical School

Arif I Dalvi, MD is a member of the following medical societies: European Neurological Society and Movement Disorders Society

Disclosure: Nothing to disclose.

Nestor Galvez-Jimenez, MD, MSc, MHA Chairman, Department of Neurology, Program Director, Movement Disorders, Department of Neurology, Division of Medicine, Cleveland Clinic Florida

Nestor Galvez-Jimenez, MD, MSc, MHA is a member of the following medical societies: American Academy of Neurology, American College of Physicians, and Movement Disorders Society

Disclosure: Nothing to disclose.

Francisco Talavera, PharmD, PhD Adjunct Assistant Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference

Disclosure: Medscape Reference Salary Employment

References

  1. Gilman S, Low PA, Quinn N, et al. Consensus statement on the diagnosis of multiple system atrophy. J Neurol Sci. 1999 Feb 1. 163(1):94-8. [View Abstract]
  2. Gilman S, Wenning GK, Low PA, et al. Second consensus statement on the diagnosis of multiple system atrophy. Neurology. 2008 Aug 26. 71(9):670-6. [View Abstract]
  3. Yoshida M, Sone M. [Mechanism of neuronal degeneration of multiple system atrophy]. Brain Nerve. 2009 Sep. 61(9):1051-60. [View Abstract]
  4. Ahmed Z, Asi YT, Sailer A, Lees AJ, Houlden H, Revesz T, et al. The neuropathology, pathophysiology and genetics of multiple system atrophy. Neuropathol Appl Neurobiol. 2012 Feb. 38(1):4-24. [View Abstract]
  5. Jellinger KA. Neuropathology of multiple system atrophy: New thoughts about pathogenesis. Mov Disord. 2014 Oct 9. [View Abstract]
  6. Jellinger KA. Neuropathological spectrum of synucleinopathies. Mov Disord. 2003 Sep. 18 Suppl 6:S2-12. [View Abstract]
  7. Nagaishi M, Yokoo H, Nakazato Y. Tau-positive glial cytoplasmic granules in multiple system atrophy. Neuropathology. 2011 Jun. 31(3):299-305. [View Abstract]
  8. Jellinger KA. Neuropathology of multiple system atrophy: new thoughts about pathogenesis. Mov Disord. 2014 Dec. 29(14):1720-41. [View Abstract]
  9. Chrysostome V, Tison F, Yekhlef F, Sourgen C, Baldi I, Dartigues JF. Epidemiology of multiple system atrophy: a prevalence and pilot risk factor study in Aquitaine, France. Neuroepidemiology. 2004 Jul-Aug. 23(4):201-8. [View Abstract]
  10. Vanacore N. Epidemiological evidence on multiple system atrophy. J Neural Transm. 2005 Dec. 112(12):1605-12. [View Abstract]
  11. Yoshida M. [Multiple system atrophy - synuclein and neuronal degeneration]. Rinsho Shinkeigaku. 2011 Nov. 51(11):838-42. [View Abstract]
  12. Schrag A, Wenning GK, Quinn N, Ben-Shlomo Y. Survival in multiple system atrophy. Mov Disord. 2008 Jan 30. 23(2):294-6. [View Abstract]
  13. Figueroa JJ, Singer W, Parsaik A, Benarroch EE, Ahlskog JE, Fealey RD, et al. Multiple system atrophy: prognostic indicators of survival. Mov Disord. 2014 Aug. 29(9):1151-7. [View Abstract]
  14. Blumin JH, Berke GS. Bilateral vocal fold paresis and multiple system atrophy. Arch Otolaryngol Head Neck Surg. 2002 Dec. 128(12):1404-7. [View Abstract]
  15. Camacho V, Marquié M, Lleó A, et al. Cardiac sympathetic impairment parallels nigrostriatal degeneration in Probable Dementia with Lewy Bodies. Q J Nucl Med Mol Imaging. 2011 Aug. 55(4):476-83. [View Abstract]
  16. Kawai Y, Suenaga M, Takeda A, et al. Cognitive impairments in multiple system atrophy: MSA-C vs MSA-P. Neurology. 2008 Apr 15. 70(16 Pt 2):1390-6. [View Abstract]
  17. Klein C, Brown R, Wenning G, Quinn N. The "cold hands sign" in multiple system atrophy. Mov Disord. 1997 Jul. 12(4):514-8. [View Abstract]
  18. Pellecchia MT, Pivonello R, Colao A, Barone P. Growth hormone stimulation tests in the differential diagnosis of Parkinson's disease. Clin Med Res. 2006 Dec. 4(4):322-5. [View Abstract]
  19. Kimber JR, Watson L, Mathias CJ. Distinction of idiopathic Parkinson's disease from multiple-system atrophy by stimulation of growth-hormone release with clonidine. Lancet. 1997 Jun 28. 349(9069):1877-81. [View Abstract]
  20. Santamaria J, Iranzo A. Multiple System Atrophy and Sleep. Sleep Med Clin. 2008. 3:337-345.
  21. Abbott SM, Videnovic A. Sleep Disorders in Atypical Parkinsonism. Mov Disord Clin Pract (Hoboken). 2014 Jun 1. 1(2):89-96. [View Abstract]
  22. Yoshida M. Multiple system atrophy -synuclein and neuronal degeneration. Rinsho Shinkeigaku. 2011 Nov. 51(11):838-42. [View Abstract]
  23. Treglia G, Stefanelli A, Cason E, Cocciolillo F, Di Giuda D, Giordano A. Diagnostic performance of iodine-123-metaiodobenzylguanidine scintigraphy in differential diagnosis between Parkinson's disease and multiple-system atrophy: a systematic review and a meta-analysis. Clin Neurol Neurosurg. 2011 Dec. 113(10):823-9. [View Abstract]
  24. Haga R, Sugimoto K, Nishijima H, Miki Y, Suzuki C, Wakabayashi K, et al. Clinical Utility of Skin Biopsy in Differentiating between Parkinson's Disease and Multiple System Atrophy. Parkinsons Dis. 2015. 2015:167038. [View Abstract]
  25. Massimo G, Limbucci N, Catalucci A, Massimo C. Neurodegenerative Diseases. Radiol Clin N Am. 2008. 46:799-817.
  26. Deguchi K, Ikeda K, Kume K, Takata T, Kokudo Y, Kamada M, et al. Significance of the hot-cross bun sign on T2*-weighted MRI for the diagnosis of multiple system atrophy. J Neurol. 2015 Apr 7. [View Abstract]
  27. Ghaemi M, Hilker R, Rudolf J, Sobesky J, Heiss WD. Differentiating multiple system atrophy from Parkinson's disease: contribution of striatal and midbrain MRI volumetry and multi-tracer PET imaging. J Neurol Neurosurg Psychiatry. 2002 Nov. 73(5):517-23. [View Abstract]
  28. Hauser RA, Grosset DG. [(123) I]FP-CIT (DaTscan) SPECT Brain Imaging in Patients with Suspected Parkinsonian Syndromes. J Neuroimaging. 2011 Mar 16. [View Abstract]
  29. Nissen T, Malek N, Grosset KA, Newman EJ, Patterson J, Hadley D, et al. Baseline [(123) I]FP-CIT SPECT (DaTSCAN) severity correlates with medication use at 3 years in Parkinson's disease. Acta Neurol Scand. 2014 Mar. 129(3):204-8. [View Abstract]
  30. Kuzdas-Wood D, Stefanova N, Jellinger KA, Seppi K, Schlossmacher MG, Poewe W, et al. Towards translational therapies for multiple system atrophy. Prog Neurobiol. 2014 Jul. 118:19-35. [View Abstract]
  31. Chou KL, Forman MS, Trojanowski JQ, Hurtig HI, Baltuch GH. Subthalamic nucleus deep brain stimulation in a patient with levodopa-responsive multiple system atrophy. Case report. J Neurosurg. 2004 Mar. 100(3):553-6. [View Abstract]
  32. Talmant V, Esposito P, Stilhart B, Mohr M, Tranchant C. [Subthalamic stimulation in a patient with multiple system atrophy: a clinicopathological report]. Rev Neurol (Paris). 2006 Mar. 162(3):363-70. [View Abstract]
  33. O'Sullivan SS, Massey LA, Williams DR, et al. Clinical outcomes of progressive supranuclear palsy and multiple system atrophy. Brain. 2008 May. 131:1362-72. [View Abstract]