Network


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

Hotspot


Dive into the research topics where Tohru Kitada is active.

Publication


Featured researches published by Tohru Kitada.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Loss of PINK1 causes mitochondrial functional defects and increased sensitivity to oxidative stress

Clement A. Gautier; Tohru Kitada; Jie Shen

Parkinsons disease (PD) is a common neurodegenerative disorder thought to be associated with mitochondrial dysfunction. Loss of function mutations in the putative mitochondrial protein PINK1 (PTEN-induced kinase 1) have been linked to familial forms of PD, but the relation of PINK1 to mammalian mitochondrial function remains unclear. Here, we report that germline deletion of the PINK1 gene in mice significantly impairs mitochondrial functions. Quantitative electron microscopic studies of the striatum in PINK1−/− mice at 3–4 and 24 months revealed no gross changes in the ultrastructure or the total number of mitochondria, although the number of larger mitochondria is selectively increased. Functional assays showed impaired mitochondrial respiration in the striatum but not in the cerebral cortex at 3–4 months of age, suggesting specificity of this defect for dopaminergic circuitry. Aconitase activity associated with the Krebs cycle is also reduced in the striatum of PINK1−/− mice. Interestingly, mitochondrial respiration activities in the cerebral cortex are decreased in PINK1−/− mice at 2 years compared with control mice, indicating that aging can exacerbate mitochondrial dysfunction in these mice. Furthermore, mitochondrial respiration defects can be induced in the cerebral cortex of PINK1−/− mice by cellular stress, such as exposure to H2O2 or mild heat shock. Together, our findings demonstrate that mammalian PINK1 is important for mitochondrial function and provides critical protection against both intrinsic and environmental stress, suggesting a pathogenic mechanism by which loss of PINK1 may lead to nigrostriatal degeneration in PD.


Neuron | 2005

Nigrostriatal Dopaminergic Deficits and Hypokinesia Caused by Inactivation of the Familial Parkinsonism-Linked Gene DJ-1

Matthew S. Goldberg; Antonio Pisani; Marian Haburcak; Timothy A. Vortherms; Tohru Kitada; Cinzia Costa; Youren Tong; Giuseppina Martella; Anne Tscherter; Andrea Martins; Giorgio Bernardi; Bryan L. Roth; Emmanuel N. Pothos; Paolo Calabresi; Jie Shen

The manifestations of Parkinsons disease are caused by reduced dopaminergic innervation of the striatum. Loss-of-function mutations in the DJ-1 gene cause early-onset familial parkinsonism. To investigate a possible role for DJ-1 in the dopaminergic system, we generated a mouse model bearing a germline disruption of DJ-1. Although DJ-1(-/-) mice had normal numbers of dopaminergic neurons in the substantia nigra, evoked dopamine overflow in the striatum was markedly reduced, primarily as a result of increased reuptake. Nigral neurons lacking DJ-1 were less sensitive to the inhibitory effects of D2 autoreceptor stimulation. Corticostriatal long-term potentiation was normal in medium spiny neurons of DJ-1(-/-) mice, but long-term depression (LTD) was absent. The LTD deficit was reversed by treatment with D2 but not D1 receptor agonists. Furthermore, DJ-1(-/-) mice displayed hypoactivity in the open field. Collectively, our findings suggest an essential role for DJ-1 in dopaminergic physiology and D2 receptor-mediated functions.


Proceedings of the National Academy of Sciences of the United States of America | 2007

Impaired dopamine release and synaptic plasticity in the striatum of PINK1-deficient mice

Tohru Kitada; Antonio Pisani; Douglas R. Porter; Hiroo Yamaguchi; Anne Tscherter; Giuseppina Martella; Paola Bonsi; Chen Zhang; Emmanuel N. Pothos; Jie Shen

Parkinsons disease (PD) is characterized by the selective vulnerability of the nigrostriatal dopaminergic circuit. Recently, loss-of-function mutations in the PTEN-induced kinase 1 (PINK1) gene have been linked to early-onset PD. How PINK1 deficiency causes dopaminergic dysfunction and degeneration in PD patients is unknown. Here, we investigate the physiological role of PINK1 in the nigrostriatal dopaminergic circuit through the generation and multidisciplinary analysis of PINK1−/− mutant mice. We found that numbers of dopaminergic neurons and levels of striatal dopamine (DA) and DA receptors are unchanged in PINK1−/− mice. Amperometric recordings, however, revealed decreases in evoked DA release in striatal slices and reductions in the quantal size and release frequency of catecholamine in dissociated chromaffin cells. Intracellular recordings of striatal medium spiny neurons, the major dopaminergic target, showed specific impairments of corticostriatal long-term potentiation and long-term depression in PINK1−/− mice. Consistent with a decrease in evoked DA release, these striatal plasticity impairments could be rescued by either DA receptor agonists or agents that increase DA release, such as amphetamine or l-dopa. These results reveal a critical role for PINK1 in DA release and striatal synaptic plasticity in the nigrostriatal circuit and suggest that altered dopaminergic physiology may be a pathogenic precursor to nigrostriatal degeneration.


Biochemical and Biophysical Research Communications | 2008

PINK1 controls mitochondrial localization of Parkin through direct phosphorylation

Yongsung Kim; Jeehye Park; Sunhong Kim; Saera Song; Seok-Kyu Kwon; Sang-Hee Lee; Tohru Kitada; Jin-Man Kim; Jongkyeong Chung

PTEN-induced putative kinase 1 (PINK1) and Parkin, encoded by their respective genes associated with Parkinsons disease (PD), are linked in a common pathway involved in the protection of mitochondrial integrity and function. However, the mechanism of their interaction at the biochemical level has not been investigated yet. Using both mammalian and Drosophila systems, we here demonstrate that the PINK1 kinase activity is required for its function in mitochondria. PINK1 regulates the localization of Parkin to the mitochondria in its kinase activity-dependent manner. In detail, Parkin phosphorylation by PINK1 on its linker region promotes its mitochondrial translocation, and the RING1 domain of Parkin is critical for this occurrence. These results demonstrate the biochemical relationship between PINK1, Parkin, and the mitochondria and thereby suggest the possible mechanism of PINK-Parkin-associated PD pathogenesis.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Cytoplasmic Pink1 activity protects neurons from dopaminergic neurotoxin MPTP

M. Emdadul Haque; Kelly Jean Thomas; Cheryl D'Souza; Steve Callaghan; Tohru Kitada; Ruth S. Slack; Paul D. Fraser; Mark R. Cookson; Anurag Tandon; David S. Park

PTEN-induced putative kinase 1 (Pink1) is a recently identified gene linked to a recessive form of familial Parkinsons disease (PD). The kinase contains a mitochondrial localization sequence and is reported to reside, at least in part, in mitochondria. However, neither the manner by which the loss of Pink1 contributes to dopamine neuron loss nor its impact on mitochondrial function and relevance to death is clear. Here, we report that depletion of Pink1 by RNAi increased neuronal toxicity induced by MPP+. Moreover, wild-type Pink1, but not the G309D mutant linked to familial PD or an engineered kinase-dead mutant K219M, protects neurons against MPTP both in vitro and in vivo. Intriguingly, a mutant that contains a deletion of the putative mitochondrial-targeting motif was targeted to the cytoplasm but still provided protection against 1-methyl-4-phenylpyridine (MPP+)/1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced toxicity. In addition, we also show that endogenous Pink1 is localized to cytosolic as well as mitochondrial fractions. Thus, our findings indicate that Pink1 plays a functional role in the survival of neurons and that cytoplasmic targets, in addition to its other actions in the mitochondria, may be important for this protective effect.


Journal of Neurochemistry | 2009

Absence of nigral degeneration in aged parkin/DJ‐1/PINK1 triple knockout mice

Tohru Kitada; Youren Tong; Clement A. Gautier; Jie Shen

Recessively inherited loss‐of‐function mutations in the parkin, DJ‐1, or PINK1 gene are linked to familial cases of early‐onset Parkinson’s diseases (PD), and heterozygous mutations are associated with increased incidence of late‐onset PD. We previously reported that single knockout mice lacking Parkin, DJ‐1, or PINK1 exhibited no nigral degeneration, even though evoked dopamine release from nigrostriatal terminals was reduced and striatal synaptic plasticity was impaired. In this study, we tested whether inactivation of all three recessive PD genes, each of which was required for nigral neuron survival in the aging human brain, resulted in nigral degeneration during the lifespan of mice. Surprisingly, we found that triple knockout mice lacking Parkin, DJ‐1, and PINK1 have normal morphology and numbers of dopaminergic and noradrenergic neurons in the substantia nigra and locus coeruleus, respectively, at the ages of 3, 16, and 24 months. Interestingly, levels of striatal dopamine in triple knockout mice were normal at 16 months of age but increased at 24 months. These results demonstrate that inactivation of all three recessive PD genes is insufficient to cause significant nigral degeneration within the lifespan of mice, suggesting that these genes may be protective rather than essential for the survival of dopaminergic neurons during the aging process. These findings also support the notion that mammalian Parkin and PINK1 may function in the same genetic pathway as in Drosophila.


Journal of Neurochemistry | 2009

Impaired dopamine release and synaptic plasticity in the striatum of Parkin−/− mice

Tohru Kitada; Antonio Pisani; Maha Karouani; Marian Haburcak; Giuseppina Martella; Anne Tscherter; Paola Platania; Bei Wu; Emmanuel N. Pothos; Jie Shen

Parkin is the most common causative gene of juvenile and early‐onset familial Parkinson’s diseases and is thought to function as an E3 ubiquitin ligase in the ubiquitin‐proteasome system. However, it remains unclear how loss of Parkin protein causes dopaminergic dysfunction and nigral neurodegeneration. To investigate the pathogenic mechanism underlying these mutations, we used parkin−/− mice to study its physiological function in the nigrostriatal circuit. Amperometric recordings showed decreases in evoked dopamine release in acute striatal slices of parkin−/− mice and reductions in the total catecholamine release and quantal size in dissociated chromaffin cells derived from parkin−/− mice. Intracellular recordings of striatal medium spiny neurons revealed impairments of long‐term depression and long‐term potentiation in parkin−/− mice, whereas long‐term potentiation was normal in the Schaeffer collateral pathway of the hippocampus. Levels of dopamine receptors and dopamine transporters were normal in the parkin−/− striatum. These results indicate that Parkin is involved in the regulation of evoked dopamine release and striatal synaptic plasticity in the nigrostriatal pathway, and suggest that impairment in evoked dopamine release may represent a common pathophysiological change in recessive parkinsonism.


PLOS ONE | 2012

Loss of DJ-1 Does Not Affect Mitochondrial Respiration but Increases ROS Production and Mitochondrial Permeability Transition Pore Opening

Emilie Giaime; Hiroo Yamaguchi; Clement A. Gautier; Tohru Kitada; Jie Shen

Background Loss of function mutations in the DJ-1 gene have been linked to recessively inherited forms of Parkinsonism. Mitochondrial dysfunction and increased oxidative stress are thought to be key events in the pathogenesis of Parkinson’s disease. Although it has been reported that DJ-1 serves as scavenger for reactive oxidative species (ROS) by oxidation on its cysteine residues, how loss of DJ-1 affects mitochondrial function is less clear. Methodology/Principal Findings Using primary mouse embryonic fibroblasts (MEFs) or brains from DJ-1−/− mice, we found that loss of DJ-1 does not affect mitochondrial respiration. Specifically, endogenous respiratory activity as well as basal and maximal respiration are normal in intact DJ-1−/− MEFs, and substrate-specific state 3 and state 4 mitochondrial respiration are also unaffected in permeabilized DJ-1−/− MEFs and in isolated mitochondria from the cerebral cortex of DJ-1−/− mice at 3 months or 2 years of age. Expression levels and activities of all individual complexes composing the electron transport system are unchanged, but ATP production is reduced in DJ-1−/− MEFs. Mitochondrial transmembrane potential is decreased in the absence of DJ-1. Furthermore, mitochondrial permeability transition pore opening is increased, whereas mitochondrial calcium levels are unchanged in DJ-1−/− cells. Consistent with earlier reports, production of reactive oxygen species (ROS) is increased, though levels of antioxidative enzymes are unaltered. Interestingly, the decreased mitochondrial transmembrane potential and the increased mitochondrial permeability transition pore opening in DJ-1−/− MEFs can be restored by antioxidant treatment, whereas oxidative stress inducers have the opposite effects on mitochondrial transmembrane potential and mitochondrial permeability transition pore opening. Conclusions/Significance Our study shows that loss of DJ-1 does not affect mitochondrial respiration or mitochondrial calcium levels but increases ROS production, leading to elevated mitochondrial permeability transition pore opening and reduced mitochondrial transmembrane potential.


Journal of Neuroscience Research | 2010

Parkin Reverses Intracellular β-Amyloid Accumulation and Its Negative Effects on Proteasome Function

Kenneth M. Rosen; Charbel E H Moussa; Han-Kyu Lee; Pravir Kumar; Tohru Kitada; Gangjian Qin; Qinghao Fu; Henry W. Querfurth

The significance of intracellular β‐amyloid (Aβ42) accumulation is increasingly recognized in Alzheimers disease (AD) pathogenesis. Aβ removal mechanisms that have attracted attention include IDE/neprilysin degradation and antibody‐mediated uptake by immune cells. However, the role of the ubiquitin‐proteasome system (UPS) in the disposal of cellular Aβ has not been fully explored. The E3 ubiquitin ligase Parkin targets several proteins for UPS degradation, and Parkin mutations are the major cause of autosomal recessive Parkinsons disease. We tested whether Parkin has cross‐function to target misfolded proteins in AD for proteasome‐dependent clearance in SH‐SY5Y and primary neuronal cells. Wild‐type Parkin greatly decreased steady‐state levels of intracellular Aβ42, an action abrogated by proteasome inhibitors. Intracellular Aβ42 accumulation decreased cell viability and proteasome activity. Accordingly, Parkin reversed both effects. Changes in mitochondrial ATP production from Aβ or Parkin did not account for their effects on the proteasome. Parkin knock‐down led to accumulation of Aβ. In AD brain, Parkin was found to interact with Aβ and its levels were reduced. Thus, Parkin is cytoprotective, partially by increasing the removal of cellular Aβ through a proteasome‐dependent pathway.


Journal of Biological Chemistry | 2012

Inactivation of Pink1 Gene in Vivo Sensitizes Dopamine-producing Neurons to 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and Can Be Rescued by Autosomal Recessive Parkinson Disease Genes, Parkin or DJ-1

M. Emdadul Haque; Matthew P. Mount; Farzaneh Safarpour; Elizabeth Abdel-Messih; Steve Callaghan; Chantal Mazerolle; Tohru Kitada; Ruth S. Slack; Valerie A. Wallace; Jie Shen; Hymie Anisman; David S. Park

Background: Mutations in Pink1 are associated with Parkinson disease. Results: Mouse Pink1 deficiency results in hypersensitivity to MPTP-induced dopaminergic neuronal loss, which can be rescued with expression of human Parkin or DJ-1. Conclusion: Pink1 gene can regulate response to exogenous stress. Significance: These results indicate how endogenous Pink1 plays an important role in management of exogenous stress in mouse brain. Mutations in the mitochondrial PTEN-induced kinase 1 (Pink1) gene have been linked to Parkinson disease (PD). Recent reports including our own indicated that ectopic Pink1 expression is protective against toxic insult in vitro, suggesting a potential role for endogenous Pink1 in mediating survival. However, the role of endogenous Pink1 in survival, particularly in vivo, is unclear. To address this critical question, we examined whether down-regulation of Pink1 affects dopaminergic neuron loss following 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) in the adult mouse. Two model systems were utilized: virally delivered shRNA-mediated knockdown of Pink1 and germ line-deficient mice. In both instances, loss of Pink1 generated significant sensitivity to damage induced by systemic MPTP treatment. This sensitivity was associated with greater loss of dopaminergic neurons in the Substantia Nigra pars compacta and terminal dopamine fiber density in the striatum region. Importantly, we also show that viral mediated expression of two other recessive PD-linked familial genes, DJ-1 and Parkin, can protect dopaminergic neurons even in the absence of Pink1. This evidence not only provides strong evidence for the role of endogenous Pink1 in neuronal survival, but also supports a role of DJ-1 and Parkin acting parallel or downstream of endogenous Pink1 to mediate survival in a mammalian in vivo context.

Collaboration


Dive into the Tohru Kitada's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Clement A. Gautier

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Anne Tscherter

University of Rome Tor Vergata

View shared research outputs
Top Co-Authors

Avatar

Antonio Pisani

University of Rome Tor Vergata

View shared research outputs
Top Co-Authors

Avatar

Emilie Giaime

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Giuseppina Martella

University of Rome Tor Vergata

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Youren Tong

Brigham and Women's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge