Network


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

Hotspot


Dive into the research topics where Paul T. Kotzbauer is active.

Publication


Featured researches published by Paul T. Kotzbauer.


Neuron | 1998

Persephin, a Novel Neurotrophic Factor Related to GDNF and Neurturin

Jeffrey Milbrandt; Frederic J. de Sauvage; Timothy J. Fahrner; Robert H. Baloh; Melanie L. Leitner; Malú G. Tansey; Patricia A. Lampe; Robert O. Heuckeroth; Paul T. Kotzbauer; Kelli S. Simburger; Judith P. Golden; Jamie A. Davies; Richard Vejsada; Ann C. Kato; Mary Hynes; Daniel Sherman; Merry Nishimura; Li-Chong Wang; Richard Vandlen; Barbara Moffat; Robert D. Klein; Kris Poulsen; Christa L. Gray; Alain Garces; Christopher E. Henderson; Heidi S. Phillips; Eugene M. Johnson

A novel neurotrophic factor named Persephin that is approximately 40% identical to glial cell line-derived neurotrophic factor (GDNF) and neurturin (NTN) has been identified using degenerate PCR. Persephin, like GDNF and NTN, promotes the survival of ventral midbrain dopaminergic neurons in culture and prevents their degeneration after 6-hydroxydopamine treatment in vivo. Persephin also supports the survival of motor neurons in culture and in vivo after sciatic nerve axotomy and, like GDNF, promotes ureteric bud branching. However, in contrast to GDNF and NTN, persephin does not support any of the peripheral neurons that were examined. Fibroblasts transfected with Ret and one of the coreceptors GFRalpha-1 or GFRalpha-2 do not respond to persephin, suggesting that persephin utilizes additional, or different, receptor components than GDNF and NTN.


The Journal of Neuroscience | 2008

Neuroinflammation and Oxidation/Nitration of α-Synuclein Linked to Dopaminergic Neurodegeneration

Hui-Ming Gao; Paul T. Kotzbauer; Kunihiro Uryu; Susan Leight; John Q. Trojanowski; Virginia M.-Y. Lee

α-Synuclein (SYN) is the major component of Lewy bodies, the neuropathological hallmarks of Parkinsons disease (PD). Missense mutations and multiplications of the SYN gene cause autosomal dominant inherited PD. Thus, SYN is implicated in the pathogenesis of PD. However, the mechanism whereby SYN promotes neurodegeneration remains unclear. Familial PD with SYN gene mutations are rare because the majority of PD is sporadic and emerging evidence indicates that sporadic PD may result from genetic and environmental risk factors including neuroinflammation. Hence, we examined the relationship between SYN dysfunction and neuroinflammation in mediating dopaminergic neurodegeneration in mice and dopaminergic neuronal cultures derived from wild-type SYN and mutant A53T SYN transgenic mice in a murine SYN-null (SYNKO) background (M7KO and M83KO, respectively). Stereotaxic injection of an inflammagen, lipopolysaccharide, into substantia nigra of these SYN genetically engineered mice induced similar inflammatory reactions. In M7KO and M83KO, but not in SYNKO mice, the neuroinflammation was associated with dopaminergic neuronal death and the accumulation of insoluble aggregated SYN as cytoplasmic inclusions in nigral neurons. Nitrated/oxidized SYN was detected in these inclusions and abatement of microglia-derived nitric oxide and superoxide provided significant neuroprotection in neuron–glia cultures from M7KO mice. These data suggest that nitric oxide and superoxide released by activated microglia may be mediators that link inflammation and abnormal SYN in mechanisms of PD neurodegeneration. This study advances understanding of the role of neuroinflammation and abnormal SYN in the pathogenesis of PD and opens new avenues for the discovery of more effective therapies for PD.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Heparan sulfate proteoglycans mediate internalization and propagation of specific proteopathic seeds

Brandon B. Holmes; Sarah L. DeVos; Najla Kfoury; Mei Li; Rachel Jacks; Kiran Yanamandra; Mohand Ouidir Ouidja; Frances M. Brodsky; Jayne Marasa; Devika P. Bagchi; Paul T. Kotzbauer; Timothy M. Miller; Dulce Papy-Garcia; Marc I. Diamond

Significance Prion-like propagation of proteopathic seeds may underlie the progression of neurodegenerative diseases, including the tauopathies and synucleinopathies. Aggregate entry into the cell is a crucial step in transcellular propagation. We used chemical, enzymatic, and genetic methods to identify heparan sulfate proteoglycans as critical mediators of tau aggregate binding and uptake, and subsequent seeding of normal intracellular tau. This pathway mediates aggregate uptake in cultured cells, primary neurons, and brain. α-Synuclein fibrils use the same entry mechanism to seed intracellular aggregation, whereas huntingtin fibrils do not. This establishes the molecular basis for a key step in aggregate propagation. Recent experimental evidence suggests that transcellular propagation of fibrillar protein aggregates drives the progression of neurodegenerative diseases in a prion-like manner. This phenomenon is now well described in cell and animal models and involves the release of protein aggregates into the extracellular space. Free aggregates then enter neighboring cells to seed further fibrillization. The mechanism by which aggregated extracellular proteins such as tau and α-synuclein bind and enter cells to trigger intracellular fibril formation is unknown. Prior work indicates that prion protein aggregates bind heparan sulfate proteoglycans (HSPGs) on the cell surface to transmit pathologic processes. Here, we find that tau fibril uptake also occurs via HSPG binding. This is blocked in cultured cells and primary neurons by heparin, chlorate, heparinase, and genetic knockdown of a key HSPG synthetic enzyme, Ext1. Interference with tau binding to HSPGs prevents recombinant tau fibrils from inducing intracellular aggregation and blocks transcellular aggregate propagation. In vivo, a heparin mimetic, F6, blocks neuronal uptake of stereotactically injected tau fibrils. Finally, uptake and seeding by α-synuclein fibrils, but not huntingtin fibrils, occurs by the same mechanism as tau. This work suggests a unifying mechanism of cell uptake and propagation for tauopathy and synucleinopathy.


Neurology | 2008

Neurodegeneration associated with genetic defects in phospholipase A2

Allison Gregory; Shawn K. Westaway; I. E. Holm; Paul T. Kotzbauer; Penny Hogarth; Scott Sonek; J. C. Coryell; T. M. Nguyen; Nardo Nardocci; Giovanna Zorzi; D. Rodriguez; Isabelle Desguerre; Enrico Bertini; Alessandro Simonati; Barbara Levinson; Cristina Dias; Clara Barbot; Inês Carrilho; Manuela Santos; Ibrahim Malik; Jane Gitschier; Susan J. Hayflick

Objective: Mutations in the gene encoding phospholipase A2 group VI (PLA2G6) are associated with two childhood neurologic disorders: infantile neuroaxonal dystrophy (INAD) and idiopathic neurodegeneration with brain iron accumulation (NBIA). INAD is a severe progressive psychomotor disorder in which axonal spheroids are found in brain, spinal cord, and peripheral nerves. High globus pallidus iron is an inconsistent feature of INAD; however, it is a diagnostic criterion of NBIA, which describes a clinically and genetically heterogeneous group of disorders that share this hallmark feature. We sought to delineate the clinical, radiographic, pathologic, and genetic features of disease resulting from defective phospholipase A2. Methods: We identified 56 patients clinically diagnosed with INAD and 23 with idiopathic NBIA and screened their DNA for PLA2G6 mutations. Results: Eighty percent of patients with INAD had mutations in PLA2G6, whereas mutations were found in only 20% of those with idiopathic NBIA. All patients with two null mutations had a more severe phenotype. On MRI, nearly all mutation-positive patients had cerebellar atrophy, and half showed brain iron accumulation. We observed Lewy bodies and neurofibrillary tangles in association with PLA2G6 mutations. Conclusion: Defects in phospholipase A2 lead to a range of phenotypes. PLA2G6 mutations are associated with nearly all cases of classic infantile neuroaxonal dystrophy but a minority of cases of idiopathic neurodegeneration with brain iron accumulation, and genotype correlates with phenotype. Cerebellar atrophy predicts which patients are likely to be mutation-positive. The neuropathologic changes that are caused by defective phospholipase A2 suggest a shared pathogenesis with both Parkinson and Alzheimer diseases.


The Journal of Comparative Neurology | 1998

Expression of neurturin, GDNF, and their receptors in the adult mouse CNS

Judith P. Golden; Robert H. Baloh; Paul T. Kotzbauer; Patricia A. Lampe; Patricia A. Osborne; Jeffrey Milbrandt; Eugene M. Johnson

Neurturin (NTN) and glial cell line‐derived neurotrophic factor (GDNF) are the first two members of the GDNF family (GF) of neurotrophic factors. These two proteins are potent survival factors for several populations of central and peripheral neurons in mature and developing rodents. The receptor for these factors is a multicomponent complex that includes the RET (rearranged during transfection) tyrosine kinase receptor and one of two glycosyl phosphatidylinositol (GPI)‐linked ligand‐binding components called GDNF family receptor alphas (GFRα‐1 and GFRα‐2). We have used in situ hybridization to study the mRNA expression of NTN, GDNF, RET, GFRα‐1, and GFRα‐2 in the central nervous system (CNS) of adult mice. GF receptors are expressed in several areas in which neuronal populations known to respond to NTN and GDNF are located, including the ventral horn of the spinal cord and the compacta region of the substantia nigra. In addition, we have demonstrated receptor expression in other areas of the brain including the thalamus and hypothalamus. Neurons in these areas express GF receptors, and therefore, may respond to NTN or GDNF. NTN and GDNF are expressed in targets of neurons that express GF receptors. The pattern of GF factor and receptor expression in the adult brain suggests a role for these factors in maintaining neuronal circuits in the mature CNS. J. Comp. Neurol. 398:139–150, 1998.


American Journal of Pathology | 2008

Disrupted Membrane Homeostasis and Accumulation of Ubiquitinated Proteins in a Mouse Model of Infantile Neuroaxonal Dystrophy Caused by PLA2G6 Mutations

Ibrahim Malik; John Turk; David J. Mancuso; Laura Montier; Mary Wohltmann; David F. Wozniak; Robert E. Schmidt; Richard W. Gross; Paul T. Kotzbauer

Mutations in the PLA2G6 gene, which encodes group VIA calcium-independent phospholipase A2 (iPLA(2)beta), were recently identified in patients with infantile neuroaxonal dystrophy (INAD) and neurodegeneration with brain iron accumulation. A pathological hallmark of these childhood neurodegenerative diseases is the presence of distinctive spheroids in distal axons that contain accumulated membranes. We used iPLA(2)beta-KO mice generated by homologous recombination to investigate neurodegenerative consequences of PLA2G6 mutations. iPLA(2)beta-KO mice developed age-dependent neurological impairment that was evident in rotarod, balance, and climbing tests by 13 months of age. The primary abnormality underlying this neurological impairment was the formation of spheroids containing tubulovesicular membranes remarkably similar to human INAD. Spheroids were strongly labeled with anti-ubiquitin antibodies. Accumulation of ubiquitinated protein in spheroids was evident in some brain regions as early as 4 months of age, and the onset of motor impairment correlated with a dramatic increase in ubiquitin-positive spheroids throughout the neuropil in nearly all brain regions. Furthermore accumulating ubiquitinated proteins were observed primarily in insoluble fractions of brain tissue, implicating protein aggregation in this pathogenic process. These results indicate that loss of iPLA(2)beta causes age-dependent impairment of axonal membrane homeostasis and protein degradation pathways, leading to age-dependent neurological impairment. iPLA(2)beta-KO mice will be useful for further studies of pathogenesis and experimental interventions in INAD and neurodegeneration with brain iron accumulation.


The Journal of Neuroscience | 2005

Altered Neuronal Mitochondrial Coenzyme A Synthesis in Neurodegeneration with Brain Iron Accumulation Caused by Abnormal Processing, Stability, and Catalytic Activity of Mutant Pantothenate Kinase 2

Paul T. Kotzbauer; Adam C. Truax; John Q. Trojanowski; Virginia M.-Y. Lee

Mutations in the pantothenate kinase 2 (PANK2) gene have been identified in patients with neurodegeneration with brain iron accumulation (NBIA; formerly Hallervorden-Spatz disease). However, the mechanisms by which these mutations cause neurodegeneration are unclear, especially given the existence of multiple pantothenate kinase genes in humans and multiple PanK2 transcripts with potentially different subcellular localizations. We demonstrate that PanK2 protein is localized to mitochondria of neurons in human brain, distinguishing it from other pantothenate kinases that do not possess mitochondrial-targeting sequences. PanK2 protein translated from the most 5′ start site is sequentially cleaved at two sites by the mitochondrial processing peptidase, generating a long-lived 48 kDa mature protein identical to that found in human brain extracts. The mature protein catalyzes the initial step in coenzyme A (CoA) synthesis but displays feedback inhibition in response to species of acyl CoA rather than CoA itself. Some, but not all disease-associated point mutations result in significantly reduced catalytic activity. The most common mutation, G521R, results in marked instability of the intermediate PanK2 isoform and reduced production of the mature isoform. These results suggest that NBIA is caused by altered neuronal mitochondrial lipid metabolism caused by mutations disrupting PanK2 protein levels and catalytic activity.


Neurotoxicology | 2012

Increased risk of parkinsonism associated with welding exposure

Brad A. Racette; Susan R. Criswell; Jessica I. Lundin; Angela Hobson; Noah S. Seixas; Paul T. Kotzbauer; Bradley Evanoff; Joel S. Perlmutter; Jing Zhang; Lianne Sheppard; Harvey Checkoway

OBJECTIVE Manganese (Mn), an established neurotoxicant, is a common component of welding fume. The neurological phenotype associated with welding exposures has not been well described. Prior epidemiologic evidence linking occupational welding to parkinsonism is mixed, and remains controversial. METHODS This was a cross-sectional and nested case-control study to investigate the prevalence and phenotype of parkinsonism among 811 shipyard and fabrication welders recruited from trade unions. Two reference groups included 59 non-welder trade workers and 118 newly diagnosed, untreated idiopathic PD patients. Study subjects were examined by a movement disorders specialist using the Unified Parkinson Disease Rating Scale motor subsection 3 (UPDRS3). Parkinsonism cases were defined as welders with UPDRS3 score ≥15. Normal was defined as UPDRS3<6. Exposure was classified as intensity adjusted, cumulative years of welding. Adjusted prevalence ratios for parkinsonism were calculated in relation to quartiles of welding years. RESULTS The overall prevalence estimate of parkinsonism was 15.6% in welding exposed workers compared to 0% in the reference group. Among welders, we observed a U-shaped dose-response relation between weighted welding exposure-years and parkinsonism. UPDRS3 scores for most domains were similar between welders and newly diagnosed idiopathic Parkinson disease (PD) patients, except for greater frequency of rest tremor and asymmetry in PD patients. CONCLUSION This work-site based study among welders demonstrates a high prevalence of parkinsonism compared to nonwelding-exposed workers and a clinical phenotype that overlaps substantially with PD.


Neuron | 1994

Postnatal development of survival responsiveness in rat sympathetic neurons to leukemia inhibitory factor and ciliary neurotrophic factor

Paul T. Kotzbauer; Patricia A. Lampe; Steven Estus; Jeffrey Milbrandt; Eugene M. Johnson

Embryonic rat sympathetic neurons undergo programmed cell death upon NGF deprivation. We show that during postnatal development, these neurons acquire the ability to be supported in vitro by LIF and CNTF as well as NGF. LIF and CNTF do not promote the long-term survival of embryonic day 21 sympathetic neurons in vitro. However, after 5 days of culture in the presence of NGF, the majority of embryonic day 21 sympathetic neurons can be supported by either of these factors. Furthermore, postnatal day 6 sympathetic neurons can be immediately supported by LIF and CNTF, indicating that acquisition of survival responsiveness occurs in vivo as well as in vitro. During this period, neuronal expression of LIF and CNTF receptor mRNAs remains constant, suggesting that sympathetic neurons alter their responsiveness to LIF and CNTF by allowing additional intracellular signaling pathways to promote survival.


Journal of Biological Chemistry | 2009

Genetic Ablation of Calcium-independent Phospholipase A2γ Leads to Alterations in Hippocampal Cardiolipin Content and Molecular Species Distribution, Mitochondrial Degeneration, Autophagy, and Cognitive Dysfunction

David J. Mancuso; Paul T. Kotzbauer; David F. Wozniak; Harold F. Sims; Christopher M. Jenkins; Shaoping Guan; Xianlin Han; Kui Yang; Gang Sun; Ibrahim Malik; Sara Conyers; Karen G. Green; Robert E. Schmidt; Richard W. Gross

Genetic ablation of calcium-independent phospholipase A2γ (iPLA2γ) results in profound alterations in hippocampal phospholipid metabolism and mitochondrial phospholipid homeostasis resulting in enlarged and degenerating mitochondria leading to autophagy and cognitive dysfunction. Shotgun lipidomics demonstrated multiple alterations in hippocampal lipid metabolism in iPLA2γ−/− mice including: 1) a markedly elevated hippocampal cardiolipin content with an altered molecular species composition characterized by a shift to shorter chain length molecular species; 2) alterations in both choline and ethanolamine glycerophospholipids, including a decreased plasmenylethanolamine content; 3) increased oxidized phosphatidylethanolamine molecular species; and 4) an increased content of ceramides. Electron microscopic examination demonstrated the presence of enlarged heteromorphic lamellar structures undergoing degeneration accompanied by the presence of ubiquitin positive spheroid inclusion bodies. Purification of these enlarged heteromorphic lamellar structures by buoyant density centrifugation and subsequent SDS-PAGE and proteomics identified them as degenerating mitochondria. Collectively, these results identify the obligatory role of iPLA2γ in neuronal mitochondrial lipid metabolism and membrane structure demonstrating that iPLA2γ loss of function results in a mitochondrial neurodegenerative disorder characterized by degenerating mitochondria, autophagy, and cognitive dysfunction.

Collaboration


Dive into the Paul T. Kotzbauer's collaboration.

Top Co-Authors

Avatar

Jeffrey Milbrandt

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Patricia A. Lampe

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Eugene M. Johnson

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Dhruva Dhavale

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Zhude Tu

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Joel S. Perlmutter

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Robert H. Mach

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Devika P. Bagchi

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Meghan C. Campbell

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Nigel J. Cairns

Washington University in St. Louis

View shared research outputs
Researchain Logo
Decentralizing Knowledge