Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Caryl E. Sortwell is active.

Publication


Featured researches published by Caryl E. Sortwell.


Experimental Neurology | 2001

Tumor Necrosis Factor α Is Toxic to Embryonic Mesencephalic Dopamine Neurons

Susan O. McGuire; Zaodung Ling; Jack W. Lipton; Caryl E. Sortwell; Timothy J. Collier; Paul M. Carvey

Abstract Levels of the proinflammatory cytokine tumor necrosis factor α (TNFα) are increased in postmortem brain and cerebral spinal fluid from patients with Parkinsons disease (PD). This observation provides a basis for associating TNFα with neurodegeneration, but a specific toxicity in dopamine (DA) neurons has not been firmly established. Therefore, we investigated TNFα-induced toxicity in DA neurons by utilizing primary cultures of embryonic rat mesencephalon. Exposure to TNFα resulted in a dose-dependent decrease in DA neurons as evidenced by decreased numbers of tyrosine hydroxylase-immunoreactive (THir) cells. TNFα toxicity was selective for DA neurons in that neither glial cell counts nor the total number of neurons was decreased and no general cytotoxicity was evidenced by lactate dehydrogenase assay. Many of the cells which remained immunoreactive for TH had shrunken and rounded cell bodies with broken, blunted, or absent processes. However, TNFα-treated cultures also contained some THir cells which appeared to be undamaged and possibly resistant to TNFα-induced toxicity. Additionally, immunocytochemistry revealed basal expression of TNFα receptor 1 (p55, R1) and TNFα receptor 2 (p75, R2) on all cells within the mesencephalic cultures to some degree, even though only DA neurons were affected by TNFα treatment. These data strongly suggest that TNFα mediates cell death in a sensitive population of DA neurons and support the potential involvement of proinflammatory cytokines in the degeneration of DA neurons in PD.


Experimental Neurology | 2001

A clonal line of mesencephalic progenitor cells converted to dopamine neurons by hematopoietic cytokines: a source of cells for transplantation in Parkinson's disease.

Paul M. Carvey; Zaodung Ling; Caryl E. Sortwell; Mark R. Pitzer; Susan O. McGuire; Alexander Storch; Timothy J. Collier

Neural progenitor cells potentially provide a limitless, on-demand source of cells for grafting into patients with Parkinsons disease (PD) if the signals needed to control their conversion into dopamine (DA) neurons could be identified. We have recently shown that cytokines which instruct cell division and differentiation within the hematopoeitic system may provide similar functions in the central nervous system. We have shown that mitotic progenitor cells can be isolated from embryonic rat mesencephalon and that these cells respond to a combination of interleukin-1, interleukin-11, leukemia inhibitory factor, and glial cell line-derived neurotrophic factor yielding a tyrosine hydroxylase-immunoreactive (THir) phenotype in 20-25% of total cells. In the present study, 24 clonal cell lines derived from single cells of mesencephalic proliferation spheres were examined for their response to the cytokine mixture. The clone yielding the highest percentage of THir neurons (98%) was selected for further study. This clone expressed several phenotypic characteristics of DA neurons and expression of Nurr1. The response to cytokines was stable for several passages and after cryopreservation for several months. When grafted into the striatum of DA-depleted rats, these cells attenuated rotational asymmetry to the same extent as freshly harvested embryonic DA neurons. These data demonstrate that mesencephalic progenitor cells can be clonally expanded in culture and differentiated in the presence of hematopoietic cytokines to yield enriched populations of DA neurons. When transplanted, these cells provide significant functional benefit in the rat model of PD.


Annals of Neurology | 2006

Failure of proteasome inhibitor administration to provide a model of Parkinson's disease in rats and monkeys

Jeffrey H. Kordower; Nicholas M. Kanaan; Yaping Chu; Rangasamy Suresh Babu; James Stansell; Brian T. Terpstra; Caryl E. Sortwell; Kathy Steece-Collier; Timothy J. Collier

McNaught and colleagues 1 reported recently that systemic administration of proteasome inhibitors PSI (Z‐Ileu‐Glu(OtBu)‐Ala‐Leu‐CHO) or epoxomicin recapitulated many of the degenerative changes seen in Parkinsons disease including loss of striatal dopamine and cell loss in the substantia nigra, locus ceruleus, dorsal motor nucleus of the X cranial nerve, and nucleus basalis of Meynert. Intracytoplasmic inclusions resembling Lewy bodies were also described. All experiments administering PSI to rats using identical procedures and multiple attempts failed to induce any of the previously described changes. Furthermore, administration of PSI or epoxomicin to monkeys in an attempt to extend the model to a primate species failed. Currently, systemic proteasome inhibition is not a reliable model for Parkinsons disease. Ann Neurol 2006;60:264–268


Neurobiology of Disease | 2011

TRANSFER OF HOST-DERIVED ALPHA SYNUCLEIN TO GRAFTED DOPAMINERGIC NEURONS IN RAT

Jeffrey H. Kordower; Hemraj B. Dodiya; Adam M. Kordower; Brian T. Terpstra; Katrina L. Paumier; Lalitha Madhavan; Caryl E. Sortwell; Kathy Steece-Collier; Timothy J. Collier

Multiple laboratories have recently demonstrated that long-term dopaminergic transplants form Lewy bodies in patients with Parkinsons disease. Debate has arisen as to whether these Lewy bodies form from the transfer of α synuclein from the host to the graft or whether they form from intrinsic responses of the graft from being placed into what was, or became, an inflammatory focus. To test whether the former hypothesis was possible, we grafted fetal rat ventral mesencephalon into the dopamine depleted striatum of rats that had previously received 6-hydroxydopamine lesions. One month after the transplant, rats received viral over expression of human α synuclein (AAV2/6-α synuclein) or green fluorescent protein (AAV2/6-GFP) into the striatum rostral to the grafts. Care was taken to make sure that the AAV injections were sufficiently distal to the graft so no cells would be directly transfected. All rats were sacrificed five weeks after the virus injections. Double label immunohistochemistry combined with confocal microscopy revealed that a small number of grafted tyrosine hydroxylase (TH) neurons (5.7% ± 1.5% (mean ± SEM) of grafted dopamine cells) expressed host derived α synuclein but none of the grafted cells expressed host-derived GFP. The α synuclein in a few of these cells was misfolded and failed to be digested with proteinase K. These data indicate that it is possible for host derived α synuclein to transfer to grafted neurons supporting the concept that this is one possible mechanism by which grafted dopamine neurons form Lewy bodies in Parkinsons disease patients.


Experimental Neurology | 2000

Time course of apoptotic cell death within mesencephalic cell suspension grafts : Implications for improving grafted dopamine neuron survival

Caryl E. Sortwell; Mark R. Pitzer; Timothy J. Collier

The vast majority ( congruent with 90%) of embryonic mesencephalic dopamine (DA) neurons die following transplantation to the striatum. Recent reports indicate that at least a subpopulation of grafted cells undergo apoptotic cell death at early times following implantation. This study examines the temporal pattern and magnitude of apoptotic cell death following the implantation of mesencephalic cell suspension grafts. Two techniques, a modified terminal deoxynucleotide-mediated nucleotide end labeling (TUNEL) technique and cresyl violet staining, are used to assess apoptotic cell death by detection of its biochemical and morphological identifiers, respectively. Male, Fischer 344 rats were examined at 1, 4, 7, and 28 days following implantation of embryonic day 14 (E14) ventral mesencephalic cells to the DA-denervated striatum. Results indicate that the overwhelming majority of apoptotic cell death occurs within the first 7 days after transplantation. However, the impact of the apoptosis that occurs over the first week following grafting only appears to limit grafted tyrosine hydroxylase-immunoreactive (THir) neuron survival during the first 4 days. No significant differences between the survival rates of THir neurons at 4 days after grafting and at 28 days after grafting were found. Therefore, it appears that the critical interval during which an estimated 90% of grafted DA neurons die is during the first 4 days postimplantation and that a major contributor to this cell death is apoptosis.


Neurobiology of Disease | 2006

Focal not widespread grafts induce novel dyskinetic behavior in parkinsonian rats

Eleonora Maries; Jeffrey H. Kordower; Yaping Chu; Timothy J. Collier; Caryl E. Sortwell; Eliza Olaru; Kathleen M. Shannon; Kathy Steece-Collier

Dyskinesias are a common consequence of dopaminergic therapy in patients with Parkinsons disease. Little is known about the influence of cellular replacement strategies upon drug-induced dyskinesias. In the current study, we employed parkinsonian rats to test whether the distribution of dopamine neuron grafts could differentially alter striatal circuitry and levodopa-induced dyskinesias. Specifically, we compared behavioral and neurochemical consequences of dopamine reinnervation restricted to a focal region of the striatum to innervation encompassing the majority of the striatum by distributing the same number of cells into single locus or multiple locations. Both the single-site and widespread grafts reduced pregraft dyskinesias and normalized FosB/DeltaFosB in the dorsal two-thirds of the lateral striatum. However, single-site DA graft recipients developed a robust, novel forelimb-facial stereotypy and upregulated FosB/DeltaFosB expression in the ventrolateral striatum, an area associated with movements of tongue and forelimbs. The onset of forelimb-facial stereotypy correlated with measures of increased graft function.


Neurobiology of Disease | 2015

Intrastriatal injection of pre-formed mouse α-synuclein fibrils into rats triggers α-synuclein pathology and bilateral nigrostriatal degeneration

Katrina L. Paumier; Kelvin C. Luk; Fredric P. Manfredsson; Nicholas M. Kanaan; Jack W. Lipton; Timothy J. Collier; Kathy Steece-Collier; Christopher J. Kemp; Stephanie L Celano; Emily Schulz; Ivette M. Sandoval; Sheila M. Fleming; Elliott Dirr; Nicole K. Polinski; John Q. Trojanowski; Virginia M.-Y. Lee; Caryl E. Sortwell

Previous studies demonstrate that intrastriatal injections of fibrillar alpha-synuclein (α-syn) into mice induce Parkinsons disease (PD)-like Lewy body (LB) pathology formed by aggregated α-syn in anatomically interconnected regions and significant nigrostriatal degeneration. The aim of the current study was to evaluate whether exogenous mouse α-syn pre-formed fibrils (PFF) injected into the striatum of rats would result in accumulation of LB-like intracellular inclusions and nigrostriatal degeneration. Sprague-Dawley rats received unilateral intrastriatal injections of either non-fibrillized recombinant α-syn or PFF mouse α-syn in 1- or 2- sites and were euthanized at 30, 60 or 180 days post-injection (pi). Both non-fibrillized recombinant α-syn and PFF α-syn injections resulted in phosphorylated α-syn intraneuronal accumulations (i.e., diffuse Lewy neurite (LN)- and LB-like inclusions) with significantly greater accumulations following PFF injection. LB-like inclusions were observed in several areas that innervate the striatum, most prominently the frontal and insular cortices, the amygdala, and the substantia nigra pars compacta (SNpc). α-Syn accumulations co-localized with ubiquitin, p62, and were thioflavin-S-positive and proteinase-k resistant, suggesting that PFF-induced pathology exhibits properties similar to human LBs. Although α-syn inclusions within the SNpc remained ipsilateral to striatal injection, we observed bilateral reductions in nigral dopamine neurons at the 180-day time-point in both the 1- and 2-site PFF injection paradigms. PFF injected rats exhibited bilateral reductions in striatal dopaminergic innervation at 60 and 180 days and bilateral decreases in homovanillic acid; however, dopamine reduction was observed only in the striatum ipsilateral to PFF injection. Although the level of dopamine asymmetry in PFF injected rats at 180 days was insufficient to elicit motor deficits in amphetamine-induced rotations or forelimb use in the cylinder task, significant disruption of ultrasonic vocalizations was observed. Taken together, our findings demonstrate that α-syn PFF are sufficient to seed the pathological conversion and propagation of endogenous α-syn to induce a progressive, neurodegenerative model of α-synucleinopathy in rats.


Drugs & Aging | 1999

Therapeutic Potential of Nerve Growth Factors in Parkinson’s Disease

Timothy J. Collier; Caryl E. Sortwell

Parkinson’s disease (PD) is a neurodegenerative syndrome which primarily affects dopamine-producing neurons of the substantia nigra, resulting in poverty and slowness of movement, instability of gait and posture, and tremor at rest in individuals with the disease. While symptoms of the disease can be effectively managed for several years with available drugs, the syndrome is progressive and the efficacy of standard drugs wanes with time. One experimental approach to therapy is to use natural and synthetic molecules which promote survival and growth of dopaminergic neurons, so-called ‘neurotrophic factors’, to stabilise the diminishing population of dopaminergic neurons and stimulate compensation and growth in these cells. In this review, we examine the available evidence on 29 molecules with neurotrophic properties for dopaminergic neurons. The properties of these molecules provide ample reasons for optimism that a neurotrophic strategy can be developed that would provide a significant treatment option for patients with PD. While the search continues for even more specific, potent and long-lasting agents, the single greatest challenge is the development of techniques for targeted delivery of these molecules.


Neurobiology of Disease | 2010

Stimulation of the Rat Subthalamic Nucleus is Neuroprotective Following Significant Nigral Dopamine Neuron Loss

Anne L. Spieles-Engemann; Michael M. Behbehani; Timothy J. Collier; S.L. Wohlgenant; Kathy Steece-Collier; Katrina L. Paumier; Brian F. Daley; Sara E. Gombash; Lalitha Madhavan; George T. Mandybur; Jack W. Lipton; Brian T. Terpstra; Caryl E. Sortwell

Deep brain stimulation of the subthalamic nucleus (STN-DBS) is efficacious in treating the motor symptoms of Parkinsons disease (PD). However, the impact of STN-DBS on the progression of PD is unknown. Previous preclinical studies have demonstrated that STN-DBS can attenuate the degeneration of a relatively intact nigrostriatal system from dopamine (DA)-depleting neurotoxins. The present study examined whether STN-DBS can provide neuroprotection in the face of prior significant nigral DA neuron loss similar to PD patients at the time of diagnosis. STN-DBS between 2 and 4 weeks after intrastriatal 6-hydroxydopamine (6-OHDA) provided significant sparing of DA neurons in the SN of rats. This effect was not due to inadvertent lesioning of the STN and was dependent upon proper electrode placement. Since STN-DBS appears to have significant neuroprotective properties, initiation of STN-DBS earlier in the course of PD may provide added neuroprotective benefits in addition to its ability to provide symptomatic relief.


The Journal of Comparative Neurology | 2000

Cyclosporin A protects striatal neurons in vitro and in vivo from 3-nitropropionic acid toxicity

Liza Leventhal; Caryl E. Sortwell; Rose Hanbury; Timothy J. Collier; Jeffrey H. Kordower; Stéphane Palfi

The neuroprotective properties of cyclosporin A (CsA) are mediated by its ability to prevent mitochondrial permeability transition during exposure to high levels of calcium or oxidative stress. By using the mitochondrial toxin 3‐nitropropionic acid (3NP), the present study assessed whether CsA could protect striatal neurons in vitro and in vivo. In vitro, 3NP produced a 20–30% reduction of striatal glutamic acid decarboxylase‐immunoreactive (GAD‐ir) neurons. A single treatment with CsA protected GAD‐ir neurons from 3NP toxicity at lower (0.2 or 1.0 μM), but not at higher (5.0 μM) doses. Similar findings were seen when the cultures were treated twice with cyclosporin. In vivo experiments used the Lewis rat model of Huntingtons disease (HD) in which a low 3NP dose was delivered subcutaneously through an osmotic minipump. Rats received unilateral or bilateral intrastriatal saline injections to disrupt the blood‐brain barrier (BBB) and facilitate CsA reaching vulnerable neurons. In the first experiment, CsA treated 3NP‐lesioned rats displayed significantly more dopamine‐and adenosine‐3`,5`‐monophosphate‐regulated phosphoprotein (DARPP32‐ir) neurons ipsilateral to BBB disruption compared to the contralateral intact striatum, indicating that disruption of the BBB maybe necessary for CsAs neuroprotective effects. In the second experiment, stereological counts of DARPP32‐ir neurons revealed that CsA protected striatal neurons in a dose‐dependent manner following bilateral disruption of the striatal BBB. Rats treated with the higher (15 or 20 mg/kg) but not lower (5 mg/kg) doses of CsA displayed greater numbers of DARRP32‐ir striatal neurons relative to vehicle‐treated 3NP‐lesioned rats. Thus, under conditions in which CsA can gain access to striatal neurons, significant protection from 3NP toxicity is observed. Therefore, CsA or more lipophilic analogues of this compound, may be of potential therapeutic benefit by protecting vulnerable neurons from the primary pathological event observed in HD. J. Comp. Neurol. 425:471–478, 2000.

Collaboration


Dive into the Caryl E. Sortwell's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jack W. Lipton

Michigan State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

D. Luke Fischer

Michigan State University

View shared research outputs
Top Co-Authors

Avatar

Susan O. McGuire

Rush University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge