Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Cancer Immunol Res ; 2024 Mar 20.
Article in English | MEDLINE | ID: mdl-38558120

ABSTRACT

Small cell lung cancer (SCLC) is an aggressive cancer for which immune checkpoint inhibitors (ICIs) have had only limited success. Bispecific T-cell engagers are promising therapeutic alternatives for ICI-resistant tumors, but not all SCLC patients are responsive. Herein, to integrate CD137 costimulatory function into a T-cell engager format and thereby augment therapeutic efficacy, we generated a CD3/CD137 dual-specific Fab and engineered a DLL3-targeted trispecific antibody (DLL3 trispecific). The CD3/CD137 dual-specific Fab was generated to competitively bind to CD3 and CD137 to prevent DLL3-independent cross-linking of CD3 and CD137, which could lead to systemic T-cell activation. We demonstrated that DLL3 trispecific induced better tumor growth control and a marked increase in the number of intratumoral T cells compared to a conventional DLL3-targeted bispecific T-cell engager. These findings suggest that DLL3 trispecific can exert potent efficacy by inducing concurrent CD137 costimulation and provide a promising therapeutic option for SCLC.

2.
Cancer Immunol Res ; : OF1-OF12, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38563577

ABSTRACT

Small-cell lung cancer (SCLC) is an aggressive cancer for which immune checkpoint inhibitors (ICI) have had only limited success. Bispecific T-cell engagers are promising therapeutic alternatives for ICI-resistant tumors, but not all patients with SCLC are responsive. Herein, to integrate CD137 costimulatory function into a T-cell engager format and thereby augment therapeutic efficacy, we generated a CD3/CD137 dual-specific Fab and engineered a DLL3-targeted trispecific antibody (DLL3 trispecific). The CD3/CD137 dual-specific Fab was generated to competitively bind to CD3 and CD137 to prevent DLL3-independent cross-linking of CD3 and CD137, which could lead to systemic T-cell activation. We demonstrated that DLL3 trispecific induced better tumor growth control and a marked increase in the number of intratumoral T cells compared with a conventional DLL3-targeted bispecific T-cell engager. These findings suggest that DLL3 trispecific can exert potent efficacy by inducing concurrent CD137 costimulation and provide a promising therapeutic option for SCLC.

3.
Biomolecules ; 14(1)2024 Jan 18.
Article in English | MEDLINE | ID: mdl-38254727

ABSTRACT

Notch signaling is conserved in C. elegans, Drosophila, and mammals. Among the four NOTCH genes in humans, NOTCH1, NOTCH2, and NOTCH3 are known to cause monogenic hereditary disorders. Most NOTCH-related disorders are congenital and caused by a gain or loss of Notch signaling activity. In contrast, cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) caused by NOTCH3 is adult-onset and considered to be caused by accumulation of the mutant NOTCH3 extracellular domain (N3ECD) and, possibly, by an impairment in Notch signaling. Pathophysiological processes following mutant N3ECD accumulation have been intensively investigated; however, the process leading to N3ECD accumulation and its association with canonical NOTCH3 signaling remain unknown. We reviewed the progress in clarifying the pathophysiological process involving mutant NOTCH3.


Subject(s)
CADASIL , Cerebral Small Vessel Diseases , Adult , Humans , Animals , CADASIL/genetics , Caenorhabditis elegans , Signal Transduction/genetics , Mutation , Drosophila , Mammals , Receptor, Notch3/genetics
4.
Nat Commun ; 13(1): 5265, 2022 09 07.
Article in English | MEDLINE | ID: mdl-36071036

ABSTRACT

Identifying a strategy with strong efficacy against non-inflamed tumours is vital in cancer immune therapy. ERY974 is a humanized IgG4 bispecific T cell-redirecting antibody that recognizes glypican-3 and CD3. Here we examine the combination effect of ERY974 and chemotherapy (paclitaxel, cisplatin, and capecitabine) in the treatment of non-inflamed tumours in a xenograft model. ERY974 monotherapy shows a minor antitumour effect on non-inflamed NCI-H446 xenografted tumours, as infiltration of ERY974-redirected T cells is limited to the tumour-stromal boundary. However, combination therapy improves efficacy by promoting T cell infiltration into the tumour centre, and increasing ERY974 distribution in the tumour. ERY974 increases capecitabine-induced cytotoxicity by promoting capecitabine conversion to its active form by inducing thymidine phosphorylase expression in non-inflamed MKN45 tumour through ERY974-induced IFNγ and TNFα in T cells. We show that ERY974 with chemotherapy synergistically and reciprocally increases antitumour efficacy, eradicating non-inflamed tumours.


Subject(s)
Antibodies, Bispecific , Antineoplastic Agents , Neoplasms , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , Antineoplastic Agents/pharmacology , Capecitabine , Humans , Neoplasms/drug therapy , T-Lymphocytes
5.
Cell Rep ; 33(12): 108542, 2020 12 22.
Article in English | MEDLINE | ID: mdl-33357423

ABSTRACT

The extracellular adenosine triphosphate (ATP) concentration is highly elevated in the tumor microenvironment (TME) and remains tightly regulated in normal tissues. Using phage display technology, we establish a method to identify an antibody that can bind to an antigen only in the presence of ATP. Crystallography analysis reveals that ATP bound in between the antibody-antigen interface serves as a switch for antigen binding. In a transgenic mouse model overexpressing the antigen systemically, the ATP switch antibody binds to the antigen in tumors with minimal binding in normal tissues and plasma and inhibits tumor growth. Thus, we demonstrate that elevated extracellular ATP concentration can be exploited to specifically target the TME, giving therapeutic antibodies the ability to overcome on-target off-tumor toxicity.


Subject(s)
Adenosine Triphosphate/metabolism , Antibodies/metabolism , Extracellular Space/metabolism , Animals , Humans , Mice , Tumor Microenvironment
6.
Neurochem Int ; 139: 104816, 2020 10.
Article in English | MEDLINE | ID: mdl-32758590

ABSTRACT

Patients with Parkinson's disease (PD) show a common progressive neurodegenerative movement disorder characterized by rigidity, tremors, postural instability, and bradykinesia due to the loss of dopaminergic neurons in the substantia nigra, and is often accompanied by several non-motor symptoms, called parkinsonism. Several lines of recent evidence support the hypothesis that mutations in the gene encoding phosphoglycerate kinase (PGK) play an important role in the PD mechanism. PGK is a key enzyme in the glycolytic pathway that catalyzes the reaction from 1,3-diphosphoglycerate to 3-phosphoglycerate. We herein established a parkinsonism model targeting Drosophila Pgk. Dopaminergic (DA) neuron-specific Pgk knockdown lead to locomotive defects in both young and aged adult flies and was accompanied by progressive DA neuron loss with aging. Pgk knockdown in DA neurons decreased dopamine levels in the central nervous system (CNS) of both young and aged adult flies. These phenotypes are similar to the defects observed in human PD patients, suggesting that the Pgk knockdown flies established herein are a promising model for parkinsonism. Furthermore, pan-neuron-specific Pgk knockdown induced low ATP levels and the accumulation of reactive oxygen species (ROS) in the CNS of third instar larvae. Collectively, these results indicate that a failure in the energy production system of Pgk knockdown flies causes locomotive defects accompanied by neuronal dysfunction and degeneration in DA neurons.


Subject(s)
Dopaminergic Neurons/enzymology , Parkinsonian Disorders/enzymology , Parkinsonian Disorders/genetics , Phosphoglycerate Kinase/antagonists & inhibitors , Phosphoglycerate Kinase/genetics , Amino Acid Sequence , Animals , Animals, Genetically Modified , Dopaminergic Neurons/pathology , Drosophila , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/deficiency , Drosophila Proteins/genetics , Humans , Parkinsonian Disorders/pathology , Phosphoglycerate Kinase/deficiency
7.
Front Biosci (Landmark Ed) ; 24(7): 1241-1258, 2019 06 01.
Article in English | MEDLINE | ID: mdl-31136977

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder that is characterized by the progressive degeneration of both upper motor neurons in the motor cortex and lower motor neurons in the brainstem and spinal cord. Recent advances in human genetics have identified more than 30 ALS-causing genes or genetic loci that include the fused in sarcoma (FUS) gene. In addition, a set of studies suggested a mutual relationship between cancer and ALS. The hpo gene, Drosophila MST was newly identified as a novel genetic modifier of the cabeza (caz), Drosophila FUS. The Hippo pathway negatively regulates the control of organ growth and tumor suppression. Moreover, the p53 tumor suppressor was found to genetically interact with caz. Frontotemporal lobar degeneration (FTLD) is characterized by the degeneration of neurons in the frontal and temporal lobes, and consists of a spectrum with ALS. Fusion protein nucleophosmin-human myeloid leukemia factor 1 (NPM-hMLF1), which is associated with the pathologies of myelodysplastic syndrome and acute myeloid leukemia, was recently shown to suppress defects in the Drosophila FTLD model expressing the human FUS gene. Further studies in the field are expected to elucidate epidemiological, genetic, and histopathological links between cancer and ALS/FTLD, and will lead to the development of therapeutic strategies. We herein summarize previous and current findings that support mutual links between cancer and ALS/FTLD.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Frontotemporal Lobar Degeneration/genetics , Motor Neurons/metabolism , Neoplasms/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Disease Models, Animal , Drosophila Proteins/genetics , Frontotemporal Lobar Degeneration/pathology , Hippo Signaling Pathway , Humans , Motor Neurons/pathology , Neoplasms/pathology , Protein Serine-Threonine Kinases/genetics , RNA-Binding Protein FUS/genetics
8.
Brain Res ; 1708: 207-219, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30578769

ABSTRACT

piRNAs, small non-coding RNAs, were considered to be restricted to germline cells. Although they have recently been detected in somatic cells including neurons, it remains unclear how piRNA biogenesis is involved in neuronal diseases. We herein examined the possible roles of Aubergine (Aub), a Piwi-family protein (PIWI) responsible for piRNA biogenesis, in the neuronal disorders, using the Cabeza (Caz) knockdown Drosophila. Caz is a Drosophila homologue of FUS, which is one of the genes causing amyotrophic lateral sclerosis (ALS). Aub overexpression enhanced the mobility defects accompanied by anatomical defects in motoneurons at neuromuscular junctions induced by the neuron-specific knockdown of Caz. In order to elucidate the underlying mechanisms, we examined pre-piRNA and mature-size piRNA levels under these conditions. qRT-PCR and RNA-seq analyses revealed that the Caz knockdown increased pre-piRNA levels, but reduced mature-size piRNA levels in the central nervous system (CNS), suggesting a role in the pre-piRNAs production. Aub overexpression did not increase mature-size piRNA levels. These results suggest that the accumulated pre-piRNAs are abnormal abortive pre-piRNAs that cannot be further processed by slicers, including Aub. We also demonstrated a relationship between Caz and pre-piRNAs in the CNS by RNA immunoprecipitation. Aub overexpression induced the abnormal cytoplasmic localization of Caz. Based on these results, we propose a model in which Caz knockdown-induced abnormal pre-piRNAs associate with Caz, then translocate and accumulate in the cytoplasm, a process that may be mediated by Aub. The novel roles for Caz and Aub demonstrated herein using the Caz-knockdown fly will contribute to a deeper understanding of the pathogenesis of ALS.


Subject(s)
Drosophila Proteins/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group F-H/metabolism , Peptide Initiation Factors/metabolism , RNA, Small Interfering/biosynthesis , Animals , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Drosophila melanogaster/metabolism , Male , Motor Neurons/metabolism , Nervous System Diseases/genetics , Nervous System Diseases/metabolism , RNA Processing, Post-Transcriptional , RNA, Small Interfering/genetics , RNA-Binding Proteins/metabolism , Transcription Factor TFIID/metabolism
9.
Exp Cell Res ; 371(2): 311-321, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30092221

ABSTRACT

Mutations in the Fused in Sarcoma (FUS) gene have been identified in familial ALS in human. Drosophila contains a single ortholog of human FUS called Cabeza (Caz). We previously established Drosophila models of ALS targeted to Caz, which developed the locomotive dysfunction and caused anatomical defects in presynaptic terminals of motoneurons. Accumulating evidence suggests that ALS and cancer share defects in many cellular processes. The Hippo pathway was originally discovered in Drosophila and plays a role as a tumor suppressor in mammals. We aimed to determine whether Hippo pathway genes modify the ALS phenotype using Caz knockdown flies. We found a genetic link between Caz and Hippo (hpo), the Drosophila ortholog of human Mammalian sterile 20-like kinase (MST) 1 and 2. Loss-of-function mutations of hpo rescued Caz knockdown-induced eye- and neuron-specific defects. The decreased Caz levels in nuclei induced by Caz knockdown were also rescued by loss of function mutations of hpo. Moreover, hpo mRNA level was dramatically increased in Caz knockdown larvae, indicating that Caz negatively regulated hpo. Our results demonstrate that hpo, Drosophila MST, is a novel modifier of Drosophila FUS. Therapeutic targets that inhibit the function of MST could modify the pathogenic processes of ALS.


Subject(s)
Cell Cycle Proteins/genetics , Cytoskeletal Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Intracellular Signaling Peptides and Proteins/genetics , Larva/genetics , Neurogenesis/genetics , Protein Serine-Threonine Kinases/genetics , RNA-Binding Proteins/genetics , Transcription Factor TFIID/genetics , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Cell Cycle Proteins/metabolism , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Drosophila Proteins/deficiency , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Eye/metabolism , Eye/ultrastructure , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Larva/cytology , Larva/growth & development , Larva/metabolism , Motor Neurons/metabolism , Motor Neurons/pathology , Nerve Degeneration , Presynaptic Terminals/metabolism , Presynaptic Terminals/ultrastructure , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Transcription Factor TFIID/deficiency
10.
Exp Neurol ; 310: 1-13, 2018 12.
Article in English | MEDLINE | ID: mdl-30165075

ABSTRACT

Neuron-specific knockdown of the dFIG4 gene, a Drosophila homologue of human FIG4 and one of the causative genes for Charcot-Marie-Tooth disease (CMT), reduces the locomotive abilities of adult flies, as well as causing defects at neuromuscular junctions, such as reduced synaptic branch length in presynaptic terminals of the motor neurons in third instar larvae. Eye imaginal disc-specific knockdown of dFIG4 induces abnormal morphology of the adult compound eye, the rough eye phenotype. In this study, we carried out modifier screening of the dFIG4 knockdown-induced rough eye phenotype using a set of chromosomal deficiency lines on the second chromosome. By genetic screening, we detected 9 and 15 chromosomal regions whose deletions either suppressed or enhanced the rough eye phenotype induced by the dFIG4 knockdown. By further genetic screening with mutants of individual genes in one of these chromosomal regions, we identified the gene CR18854 that suppressed the rough eye phenotype and the loss-of-cone cell phenotype. The CR18854 gene encodes a long non-coding RNA (lncRNA) consisting of 2566 bases. Mutation and knockdown of CR18854 patially suppressed the enlarged lysosome phenotype induced by Fat body-specific knockdown of dFIG4. Further characterization of CR18854, and a few other lncRNAs in relation to dFIG4 in neuron, using neuron-specific dFIG4 knockdown flies indicated a genetic link between the dFIG4 gene and lncRNAs including CR18854 and hsrω. We also obtained data indicating genetic interaction between CR18854 and Cabeza, a Drosophila homologue of human FUS, which is one of the causing genes for amyotrophic lateral sclerosis (ALS). These results suggest that lncRNAs such as CR18854 and hsrω are involved in a common pathway in CMT and ALS pathogenesis.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Epistasis, Genetic/genetics , Flavoproteins/genetics , Genetic Testing , Mutation/genetics , Phosphoric Monoester Hydrolases/genetics , RNA, Long Noncoding/genetics , Animals , Animals, Genetically Modified , Disease Models, Animal , Drosophila , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Eye/ultrastructure , Flavoproteins/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Lysosomes/genetics , Lysosomes/ultrastructure , Microscopy, Electron, Scanning , Movement/physiology , Neuromuscular Junction/genetics , Neuromuscular Junction/ultrastructure , Neurons/physiology , Neurons/ultrastructure , Phosphoric Monoester Hydrolases/metabolism , Pupa/genetics , RNA, Long Noncoding/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retina/cytology
11.
Sci Rep ; 8(1): 11291, 2018 07 26.
Article in English | MEDLINE | ID: mdl-30050143

ABSTRACT

Fused in sarcoma (FUS) was identified as a component of typical inclusions in frontotemporal lobar degeneration (FTLD) and amyotrophic lateral sclerosis (ALS). In FTLD, both nuclear and cytoplasmic inclusions with wild-type FUS exist, while cytoplasmic inclusions with a mutant-form of FUS occur in many ALS cases. These observations imply that FUS plays a role across these two diseases. In this study, we examined the effect of several proteins including molecular chaperons on the aberrant eye morphology phenotype induced by overexpression of wild-type human FUS (hFUS) in Drosophila eye imaginal discs. By screening, we found that the co-expression of nucleophosmin-human myeloid leukemia factor 1 (NPM-hMLF1) fusion protein could suppress the aberrant eye morphology phenotype induced by hFUS. The driving of hFUS expression at 28 °C down-regulated levels of hFUS and endogenous cabeza, a Drosophila homolog of hFUS. The down-regulation was mediated by proteasome dependent degradation. Co-expression of NPM-hMLF1 suppressed this down-regulation. In addition, co-expression of NPM-hMLF1 partially rescued pharate adult lethal phenotype induced by hFUS in motor neurons. These findings with a Drosophila model that mimics FTLD provide clues for the development of novel FTLD therapies.


Subject(s)
Animals, Genetically Modified , Frontotemporal Lobar Degeneration/pathology , Gene Expression , Nuclear Proteins/metabolism , Proteins/metabolism , RNA-Binding Protein FUS/metabolism , Recombinant Fusion Proteins/metabolism , Animals , Cell Cycle Proteins , DNA-Binding Proteins , Disease Models, Animal , Down-Regulation/radiation effects , Drosophila , Eye/embryology , Eye Abnormalities/prevention & control , Humans , Imaginal Discs/embryology , Nuclear Proteins/genetics , Nucleophosmin , Proteins/genetics , RNA-Binding Protein FUS/genetics , Recombinant Fusion Proteins/genetics , Temperature
12.
Adv Exp Med Biol ; 1076: 79-95, 2018.
Article in English | MEDLINE | ID: mdl-29951816

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease that affects upper and lower motor neurons in the brain and the spinal cord. Due to the progressive neurodegeneration, ALS leads to paralysis and death caused by respiratory failure 2-5 years after the onset of symptoms. There is no effective cure available. Most ALS cases are sporadic, without family history, whereas 10% of the cases are familial. Identification of variants in more than 30 different loci has provided insight into the pathogenic molecular mechanisms mediating disease pathogenesis. Studies of a Drosophila melanogaster model for each of the ALS genes can contribute to uncovering pathophysiological mechanism of ALS and finding targets of the disease-modifying therapy. In this review, we focus on three ALS-causing genes: TAR DNA-binding protein (TDP-43), fused in sarcoma/translocated in liposarcoma (FUS/TLS), and chromosome 9 open reading frame 72 (C9orf72).


Subject(s)
Amyotrophic Lateral Sclerosis , Disease Models, Animal , Drosophila melanogaster , Animals , Humans
13.
Neuroreport ; 29(10): 856-862, 2018 07 04.
Article in English | MEDLINE | ID: mdl-29742619

ABSTRACT

Charcot-Marie-Tooth disease (CMT) is the most common hereditary neuropathy, and more than 80 CMT-causing genes have been identified to date. CMT4J is caused by a loss-of-function mutation in the Factor-Induced-Gene 4 (FIG4) gene, the product of which plays important roles in endosome-lysosome homeostasis. We hypothesized that Mammalian sterile 20-like kinase (MST) 1 and 2, tumor-suppressor genes, are candidate modifiers of CMT4J. We therefore examined the interaction between dFIG4 and Hippo (hpo), Drosophila counterparts of FIG4 and MSTs, respectively, using the Drosophila CMT4J model with the knockdown of dFIG4. The loss-of-function allele of hpo improved the rough eye morphology, locomotive dysfunction accompanied by structural defects in the presynaptic terminals of motoneurons, and the enlargement of lysosomes caused by the knockdown of dFIG4. Therefore, we identified hpo as a modifier of phenotypes induced by the knockdown of dFIG4. These results in Drosophila may provide an insight into the pathogenesis of CMT4J and contribute toward the development of disease-modifying therapy for CMT. We also identified the regulation of endosome-lysosome homeostasis as a novel probable function of Hippo/MST.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Drosophila Proteins/genetics , Intracellular Signaling Peptides and Proteins/genetics , Lysosomes/genetics , Mutation/genetics , Protein Serine-Threonine Kinases/genetics , Alleles , Animals , Drosophila melanogaster , Gene Knockdown Techniques/methods , Motor Neurons/metabolism , Phenotype
14.
Brain Res ; 1689: 30-44, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29604258

ABSTRACT

Mutations in SLC25A46 gene have been identified in mitochondrial diseases that are sometimes classified as Charcot-Marie-Tooth disease type 2, optic atrophy and Leigh syndrome. Human SLC25A46 functions as a transporter across the outer mitochondrial membrane. However, it is still unknown how the neurodegeneration occurring in these diseases relates to the loss of SLC25A46 function. Drosophila has CG5755 (dSLC25A46) as a single human SLC25A46 homolog. Here we established pan-neuron specific dSLC25A46 knockdown flies, and examined their phenotypes. Neuron specific knockdown of dSLC25A46 resulted in an impaired motility in both larvae and adults. Defects at neuromuscular junctions (NMJs), such as reduced synaptic branch length, decreased number and size of bouton, reduced density and size of active zone were also observed with the dSLC25A46 knockdown flies. Mitochondrial hyperfusion in synapse at NMJ, accumulation of reactive oxygen species and reduction of ATP were also observed in the dSLC25A46 knockdown flies. These results indicate that depletion of SLC25A46 induces mitochondrial defects accompanied with aberrant morphology of motoneuron and reduction of active zone that results in defect in locomotive ability. In addition, it is known that SLC25A46 mutations in human cause optic atrophy and knockdown of dSLC25A46 induces aberrant morphology of optic stalk of photoreceptor neurons in third instar larvae. Morphology and development of optic stalk of photoreceptor neurons in Drosophila are precisely regulated via cell proliferation and migration. Immunocytochemical analyses of subcellular localization of dSLC25A46 revealed that dSLC25A46 localizes not only in mitochondria, but also in plasma membrane. These observations suggest that in addition to the role in mitochondrial function, plasma membrane-localized dSLC25A46 plays a role in cell proliferation and/or migration to control optic stalk formation. The dSLC25A46 knockdown fly thus recapitulates most of the phenotypes in mitochondrial disease patients, providing a useful tool to study these diseases.


Subject(s)
Disease Models, Animal , Drosophila , Mitochondrial Diseases , Adenosine Triphosphate/metabolism , Animals , Animals, Genetically Modified , Cell Membrane/metabolism , Cell Membrane/pathology , Central Nervous System/growth & development , Central Nervous System/metabolism , Central Nervous System/pathology , Compound Eye, Arthropod/growth & development , Compound Eye, Arthropod/metabolism , Compound Eye, Arthropod/pathology , Drosophila/genetics , Gene Knockdown Techniques , Humans , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Mitochondrial Proteins/genetics , Motor Activity/physiology , Neuromuscular Junction/metabolism , Neuromuscular Junction/pathology , Neurons/metabolism , Neurons/pathology , Phenotype , Phosphate Transport Proteins/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Salivary Glands/growth & development , Salivary Glands/metabolism , Salivary Glands/pathology , Sequence Homology, Amino Acid
15.
Am J Neurodegener Dis ; 7(1): 11-31, 2018.
Article in English | MEDLINE | ID: mdl-29531866

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a rapidly progressive neurodegenerative disease characterized by the motor neuron degeneration that eventually leads to complete paralysis and death within 2-5 years after disease onset. One of the major pathological hallmark of ALS is abnormal accumulation of inclusions containing TAR DNA-binding protein-43 (TDP-43). TDP-43 is normally found in the nucleus, but in ALS, it localizes in the cytoplasm as inclusions as well as in the nucleus. Loss of nuclear TDP-43 functions likely contributes to neurodegeneration. TBPH is the Drosophila ortholog of human TDP-43. In the present study, we confirmed that Drosophila models harboring TBPH knockdown develop locomotive deficits and degeneration of motoneurons (MNs) due to loss of its nuclear functions, recapitulating the human ALS phenotypes. We previously suggested that ter94, the Drosophila ortholog of human Valosin-containing protein (VCP), is a modulator of degeneration in MNs induced by knockdown of Caz, the Drosophila ortholog of human FUS. In this study, to determine the effects of VCP on TDP-43-assosiated ALS pathogenic processes, we examined genetic interactions between TBPH and ter94. Overexpression of ter94 suppressed the compound eye degeneration caused by TBPH knockdown and suppressed the morbid phenotypes caused by neuron-specific TBPH knockdown, such as locomotive dysfunction and degeneration of MN terminals. Further immunocytochemical analyses revealed that the suppression is caused by restoring the cytoplasmically mislocalized TBPH back to the nucleus. In consistent with these observations, a loss-of-function mutation of ter94 enhanced the compound eye degeneration caused by TBPH knockdown, and partially enhanced the locomotive dysfunction caused by TBPH knockdown. Our data demonstrated that expression levels of ter94 influenced the phenotypes caused by TBPH knockdown, and indicate that reagents that up-regulate the function of human VCP could modify MN degeneration in ALS caused by TDP-43 mislocalization.

16.
Sci Transl Med ; 9(410)2017 Oct 04.
Article in English | MEDLINE | ID: mdl-28978751

ABSTRACT

Cancer care is being revolutionized by immunotherapies such as immune checkpoint inhibitors, engineered T cell transfer, and cell vaccines. The bispecific T cell-redirecting antibody (TRAB) is one such promising immunotherapy, which can redirect T cells to tumor cells by engaging CD3 on a T cell and an antigen on a tumor cell. Because T cells can be redirected to tumor cells regardless of the specificity of T cell receptors, TRAB is considered efficacious for less immunogenic tumors lacking enough neoantigens. Its clinical efficacy has been exemplified by blinatumomab, a bispecific T cell engager targeting CD19 and CD3, which has shown marked clinical responses against hematological malignancies. However, the success of TRAB in solid tumors has been hampered by the lack of a target molecule with sufficient tumor selectivity to avoid "on-target off-tumor" toxicity. Glypican 3 (GPC3) is a highly tumor-specific antigen that is expressed during fetal development but is strictly suppressed in normal adult tissues. We developed ERY974, a whole humanized immunoglobulin G-structured TRAB harboring a common light chain, which bispecifically binds to GPC3 and CD3. Using a mouse model with reconstituted human immune cells, we revealed that ERY974 is highly effective in killing various types of tumors that have GPC3 expression comparable to that in clinical tumors. ERY974 also induced a robust antitumor efficacy even against tumors with nonimmunogenic features, which are difficult to treat by inhibiting immune checkpoints such as PD-1 (programmed cell death protein-1) and CTLA-4 (cytotoxic T lymphocyte-associated protein-4). Immune monitoring revealed that ERY974 converted the poorly inflamed tumor microenvironment to a highly inflamed microenvironment. Toxicology studies in cynomolgus monkeys showed transient cytokine elevation, but this was manageable and reversible. No organ toxicity was evident. These data provide a rationale for clinical testing of ERY974 for the treatment of patients with GPC3-positive solid tumors.


Subject(s)
Antibodies, Bispecific/therapeutic use , Glypicans/immunology , Neoplasms/immunology , Neoplasms/pathology , T-Lymphocytes/immunology , Animals , Antibodies, Bispecific/administration & dosage , Antibodies, Bispecific/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , CD3 Complex/metabolism , Cytokines/metabolism , Humans , Immunocompetence/drug effects , Injections, Intravenous , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Macaca fascicularis , Mice, Transgenic , Steroids/pharmacology , Steroids/therapeutic use , T-Lymphocytes/drug effects
17.
NPJ Parkinsons Dis ; 3: 13, 2017.
Article in English | MEDLINE | ID: mdl-28649613

ABSTRACT

Phosphoglycerate kinase 1 (PGK-1) is a glycolytic enzyme encoded by PGK-1, which maps to the X chromosome. PGK-1 deficiency causes X-linked recessive hereditary chronic hemolytic anemia, myopathy, and neurological disorders due to insufficient ATP regeneration. Early-onset parkinsonism has occasionally been reported as a neurological complication of this condition. However, heterozygous carriers of PGK-1 deficiency were thought to be neurologically asymptomatic. Here, we report a boy with PGK-1 deficiency and his mother, a carrier of a heterozygous mutation in PGK-1, both of whom presented with early-onset parkinsonism. The boy developed parkinsonism at 9 years of age. His parkinsonism partially responded to levodopa treatment. 123l-metaiodobenzylguanidine (MIBG) uptake was normal. His mother, who exhibited normal PGK-1 activity in erythrocytes, developed parkinsonism at 36 years of age. Her symptoms were undistinguishable from those of Parkinson's disease (PD), despite her normal uptake of MIBG. Neither a point mutation in nor multiplication of SNCA was found. Additionally, hotspots of LRRK2 and GBA were not mutated. To our knowledge, this report provides the first description of parkinsonism in a carrier of PGK-1 deficiency. Interestingly, PGK-1 is located within the confirmed susceptibility locus for PD known as PARK12. These observations suggest that PGK-1 mutations confer susceptibility to PD.

18.
Exp Neurol ; 277: 86-95, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26708557

ABSTRACT

Mutations in Factor-Induced-Gene 4 (FIG4) gene have been identified in Charcot-Marie-Tooth disease type 4J (CMT4J), Yunis-Varon syndrome and epilepsy with polymicrogyria. FIG4 protein regulates a cellular abundance of phosphatidylinositol 3,5-bisphosphate (PI(3,5)P2), a signaling lipid on the cytosolic surface of membranes of the late endosomal compartment. PI(3,5)P2 is required for retrograde membrane trafficking from lysosomal and late endosomal compartments to the Golgi. However, it is still unknown how the neurodegeneration that occurs in these diseases is related to the loss of FIG4 function. Drosophila has CG17840 (dFIG4) as a human FIG4 homolog. Here we specifically knocked down dFIG4 in various tissues, and investigated their phenotypes. Neuron-specific knockdown of dFIG4 resulted in axonal targeting aberrations of photoreceptor neurons, shortened presynaptic terminals of motor neurons in 3rd instar larvae and reduced climbing ability in adulthood and life span. Fat body-specific knockdown of dFIG4 resulted in enlarged lysosomes in cells that were detected by staining with LysoTracker. In addition, eye imaginal disk-specific knockdown of dFIG4 disrupted differentiation of pupal ommatidial cell types, such as cone cells and pigment cells, suggesting an additional role of dFIG4 during eye development.


Subject(s)
Axons/pathology , Eye Abnormalities/genetics , Gait Disorders, Neurologic/genetics , Gait Disorders, Neurologic/pathology , Longevity/genetics , Motor Neurons/pathology , Phosphoric Monoester Hydrolases/deficiency , Animals , Animals, Genetically Modified , Central Nervous System/metabolism , Central Nervous System/pathology , Disease Models, Animal , Drosophila , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Eye Abnormalities/metabolism , Eye Abnormalities/pathology , Flavoproteins/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Imaginal Discs/pathology , Lysosomes/pathology , Neuromuscular Junction/genetics , Neuromuscular Junction/pathology , Phosphoric Monoester Hydrolases/genetics , Photoreceptor Cells, Invertebrate/pathology , Psychomotor Disorders/genetics , Sequence Alignment
19.
Exp Cell Res ; 326(1): 36-45, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24928275

ABSTRACT

Amyotrophic Lateral Sclerosis (ALS) is a fatal neurodegenerative disease that causes progressive muscular weakness. Fused in Sarcoma (FUS) that has been identified in familial ALS is an RNA binding protein that is normally localized in the nucleus. However, its function in vivo is not fully understood. Drosophila has Cabeza (Caz) as a FUS homologue and specific knockdown of Caz in the eye imaginal disc and pupal retina using a GMR-GAL4 driver was here found to induce an abnormal morphology of the adult compound eyes, a rough eye phenotype. This was partially suppressed by expression of the apoptosis inhibitor P35. Knockdown of Caz exerted no apparent effect on differentiation of photoreceptor cells. However, immunostaining with an antibody to Cut that marks cone cells revealed fusion of these and ommatidia of pupal retinae. These results indicate that Caz knockdown induces apoptosis and also inhibits differentiation of cone cells, resulting in abnormal eye morphology in adults. Mutation in EGFR pathway-related genes, such as rhomboid-1, rhomboid-3 and mirror suppressed the rough eye phenotype induced by Caz knockdown. Moreover, the rhomboid-1 mutation rescued the fusion of cone cells and ommatidia observed in Caz knockdown flies. The results suggest that Caz negatively regulates the EGFR signaling pathway required for determination of cone cell fate in Drosophila.


Subject(s)
Animals, Genetically Modified/metabolism , Drosophila Proteins/metabolism , Drosophila/metabolism , ErbB Receptors/metabolism , Gene Expression Regulation, Developmental , RNA-Binding Protein FUS/metabolism , RNA-Binding Proteins/metabolism , Receptors, Invertebrate Peptide/metabolism , Retina/metabolism , Transcription Factor TFIID/metabolism , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/growth & development , Apoptosis , Blotting, Western , Cell Differentiation , Drosophila/genetics , Drosophila/growth & development , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , ErbB Receptors/genetics , Female , Immunoenzyme Techniques , Male , Microscopy, Electron, Scanning , RNA, Small Interfering/genetics , RNA-Binding Protein FUS/genetics , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , Receptors, Invertebrate Peptide/genetics , Retina/cytology , Signal Transduction , Transcription Factor TFIID/antagonists & inhibitors , Transcription Factor TFIID/genetics
20.
J Clin Neuromuscul Dis ; 15(4): 152-6, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24872213

ABSTRACT

We report a patient with adult-type Pompe disease treated with enzyme replacement therapy (ERT) for 5.5 years. We evaluated pulmonary function and muscle strength using 6-minute walk test, manual muscle test, and dynamometer-based measurement. The long-term ERT resulted in a substantial improvement in the pulmonary function and a possible stabilization followed by mild deterioration in muscle power measured by dynamometer and 6-minute walk test. Our data may rationalize the long-term use of ERT for adult-type Pompe disease in terms of maintaining pulmonary function.


Subject(s)
Enzyme Replacement Therapy , Glycogen Storage Disease Type II/drug therapy , Glycogen Storage Disease Type II/physiopathology , Muscle, Skeletal/physiopathology , Respiration , Adult , Disease Progression , Follow-Up Studies , Humans , Immunoglobulin G/analysis , Male , Muscle Strength , Physical Therapy Modalities , Respiratory Function Tests , alpha-Glucosidases/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...