Network


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

Hotspot


Dive into the research topics where Hai Ngu is active.

Publication


Featured researches published by Hai Ngu.


Nature | 2016

RIPK1 inhibits ZBP1-driven necroptosis during development

Kim Newton; Katherine E. Wickliffe; Allie Maltzman; Debra L. Dugger; Andreas Strasser; Victoria Pham; Jennie R. Lill; Merone Roose-Girma; Søren Warming; Margaret Solon; Hai Ngu; Joshua D. Webster; Vishva M. Dixit

Receptor-interacting protein kinase 1 (RIPK1) promotes cell survival—mice lacking RIPK1 die perinatally, exhibiting aberrant caspase-8-dependent apoptosis and mixed lineage kinase-like (MLKL)-dependent necroptosis. However, mice expressing catalytically inactive RIPK1 are viable, and an ill-defined pro-survival function for the RIPK1 scaffold has therefore been proposed. Here we show that the RIP homotypic interaction motif (RHIM) in RIPK1 prevents the RHIM-containing adaptor protein ZBP1 (Z-DNA binding protein 1; also known as DAI or DLM1) from activating RIPK3 upstream of MLKL. Ripk1RHIM/RHIM mice that expressed mutant RIPK1 with critical RHIM residues IQIG mutated to AAAA died around birth and exhibited RIPK3 autophosphorylation on Thr231 and Ser232, which is a hallmark of necroptosis, in the skin and thymus. Blocking necroptosis with catalytically inactive RIPK3(D161N), RHIM mutant RIPK3, RIPK3 deficiency, or MLKL deficiency prevented lethality in Ripk1RHIM/RHIM mice. Loss of ZBP1, which engages RIPK3 in response to certain viruses but previously had no defined role in development, also prevented perinatal lethality in Ripk1RHIM/RHIM mice. Consistent with the RHIM of RIPK1 functioning as a brake that prevents ZBP1 from engaging the RIPK3 RHIM, ZBP1 interacted with RIPK3 in Ripk1RHIM/RHIMMlkl−/− macrophages, but not in wild-type, Mlkl−/− or Ripk1RHIM/RHIMRipk3RHIM/RHIM macrophages. Collectively, these findings indicate that the RHIM of RIPK1 is critical for preventing ZBP1/RIPK3/MLKL-dependent necroptosis during development.


Journal of Experimental Medicine | 2013

Dual leucine zipper kinase is required for excitotoxicity-induced neuronal degeneration

Christine D. Pozniak; Arundhati Sengupta Ghosh; Alvin Gogineni; Jesse E. Hanson; Seung-Hye Lee; Jessica L. Larson; Hilda Solanoy; Daisy Bustos; Hong Li; Hai Ngu; Adrian M. Jubb; Gai Ayalon; Jiansheng Wu; Kimberly Scearce-Levie; Qiang Zhou; Robby M. Weimer; Donald S. Kirkpatrick; Joseph W. Lewcock

Loss of dual leucine zipper kinase results in attenuated JNK/c-Jun stress response pathway activation and reduced neuronal degeneration after kainic acid–induced excitotoxic seizures.


Science Translational Medicine | 2017

Loss of dual leucine zipper kinase signaling is protective in animal models of neurodegenerative disease

Claire E. Le Pichon; William J. Meilandt; Sara L. Dominguez; Hilda Solanoy; Han Lin; Hai Ngu; Alvin Gogineni; Arundhati Sengupta Ghosh; Zhiyu Jiang; Seung-Hye Lee; Janice Maloney; Vineela D. Gandham; Christine D. Pozniak; Bei Wang; Sebum Lee; Michael Siu; Snahel Patel; Zora Modrusan; Xingrong Liu; York Rudhard; Miriam Baca; Amy Gustafson; Josh Kaminker; Richard A. D. Carano; Eric J. Huang; Oded Foreman; Robby M. Weimer; Kimberly Scearce-Levie; Joseph W. Lewcock

Blocking dual leucine zipper kinase slows disease progression in animal models of ALS and Alzheimer’s disease. A new therapeutic target zips into view The genetics, pathology, and clinical manifestations of chronic neurodegenerative diseases, such as amyotrophic lateral sclerosis (ALS), are heterogeneous, which has made the development and testing of candidate therapeutics difficult. Here, Le Pichon et al. identify dual leucine zipper kinase (DLK) as a common regulator of neuronal degeneration in mouse models of ALS and Alzheimer’s disease and in human patient postmortem brain tissue. Deletion of DLK or treatment with a DLK inhibitor resulted in neuronal protection and slowing of disease progression after diverse insults in several mouse models of neurodegenerative disease. This suggests that DLK may have broad applicability as a therapeutic target for the treatment of a number of neurodegenerative diseases. Hallmarks of chronic neurodegenerative disease include progressive synaptic loss and neuronal cell death, yet the cellular pathways that underlie these processes remain largely undefined. We provide evidence that dual leucine zipper kinase (DLK) is an essential regulator of the progressive neurodegeneration that occurs in amyotrophic lateral sclerosis and Alzheimer’s disease. We demonstrate that DLK/c-Jun N-terminal kinase signaling was increased in mouse models and human patients with these disorders and that genetic deletion of DLK protected against axon degeneration, neuronal loss, and functional decline in vivo. Furthermore, pharmacological inhibition of DLK activity was sufficient to attenuate the neuronal stress response and to provide functional benefit even in the presence of ongoing disease. These findings demonstrate that pathological activation of DLK is a conserved mechanism that regulates neurodegeneration and suggest that DLK inhibition may be a potential approach to treat multiple neurodegenerative diseases.


PLOS ONE | 2013

EGFR Inhibitor Erlotinib Delays Disease Progression but Does Not Extend Survival in the SOD1 Mouse Model of ALS

Claire E. Le Pichon; Sara L. Dominguez; Hilda Solanoy; Hai Ngu; Nicholas Lewin-Koh; Mark J. Chen; Jeffrey Eastham-Anderson; Ryan J. Watts; Kimberly Scearce-Levie

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease that causes progressive paralysis due to motor neuron death. Several lines of published evidence suggested that inhibition of epidermal growth factor receptor (EGFR) signaling might protect neurons from degeneration. To test this hypothesis in vivo, we treated the SOD1 transgenic mouse model of ALS with erlotinib, an EGFR inhibitor clinically approved for oncology indications. Although erlotinib failed to extend ALS mouse survival it did provide a modest but significant delay in the onset of multiple behavioral measures of disease progression. However, given the lack of protection of motor neuron synapses and the lack of survival extension, the small benefits observed after erlotinib treatment appear purely symptomatic, with no modification of disease course.


eNeuro | 2015

Cognitive Deficits, Changes in Synaptic Function, and Brain Pathology in a Mouse Model of Normal Aging(1,2,3).

Martin Weber; Tiffany Wu; Jesse E. Hanson; Nazia M. Alam; Hilda Solanoy; Hai Ngu; Benjamin E. Lauffer; Han H. Lin; Sara L. Dominguez; Jens Reeder; Jennifer Tom; Pascal Steiner; Oded Foreman; Glen T. Prusky; Kimberly Scearce-Levie

Abstract Age is the main risk factor for sporadic Alzheimer’s disease. Yet, cognitive decline in aged rodents has been less well studied, possibly due to concomitant changes in sensory or locomotor function that can complicate cognitive tests. We tested mice that were 3, 11, and 23 months old in cognitive, sensory, and motor measures, and postmortem measures of gliosis and neural activity (c-Fos). Hippocampal synaptic function was also examined. While age-related impairments were detectable in tests of spatial memory, greater age-dependent effects were observed in tests of associative learning [active avoidance (AA)]. Gross visual function was largely normal, but startle responses to acoustic stimuli decreased with increased age, possibly due to hearing impairments. Therefore, a novel AA variant in which light alone served as the conditioning stimuli was used. Age-related deficits were again observed. Mild changes in vision, as measured by optokinetic responses, were detected in 19- versus 4-month-old mice, but these were not correlated to AA performance. Thus, deficits in hearing or vision are unlikely to account for the observed deficits in cognitive measures. Increased gliosis was observed in the hippocampal formation at older ages. Age-related changes in neural function and plasticity were observed with decreased c-Fos in the dentate gyrus, and decreased synaptic strength and paired-pulse facilitation in CA1 slices. This work, which carefully outlines age-dependent impairments in cognitive and synaptic function, c-Fos activity, and gliosis during normal aging in the mouse, suggests robust translational measures that will facilitate further study of the biology of aging.


JCI insight | 2016

Depletion of major pathogenic cells in asthma by targeting CRTh2

Tao Huang; Meredith Hazen; Yonglei Shang; Meijuan Zhou; Xiumin Wu; Donghong Yan; Zhonghua Lin; Margaret Solon; Elizabeth Luis; Hai Ngu; Yongchang Shi; Arna Katewa; David F. Choy; Nandhini Ramamoorthi; Erick R. Castellanos; Mercedesz Balazs; Min Xu; Wyne P. Lee; Marissa L. Matsumoto; Jian Payandeh; Joseph R. Arron; Jo-Anne Hongo; Jianyong Wang; Isidro Hotzel; Cary D. Austin; Karin Reif

Eosinophilic inflammation and Th2 cytokine production are central to the pathogenesis of asthma. Agents that target either eosinophils or single Th2 cytokines have shown benefits in subsets of biomarker-positive patients. More broadly effective treatment or disease-modifying effects may be achieved by eliminating more than one inflammatory stimulator. Here we present a strategy to concomitantly deplete Th2 T cells, eosinophils, basophils, and type-2 innate lymphoid cells (ILC2s) by generating monoclonal antibodies with enhanced effector function (19A2) that target CRTh2 present on all 4 cell types. Using human CRTh2 (hCRTh2) transgenic mice that mimic the expression pattern of hCRTh2 on innate immune cells but not Th2 cells, we demonstrate that anti-hCRTh2 antibodies specifically eliminate hCRTh2+ basophils, eosinophils, and ILC2s from lung and lymphoid organs in models of asthma and Nippostrongylus brasiliensis infection. Innate cell depletion was accompanied by a decrease of several Th2 cytokines and chemokines. hCRTh2-specific antibodies were also active on human Th2 cells in vivo in a human Th2-PBMC-SCID mouse model. We developed humanized hCRTh2-specific antibodies that potently induce antibody-dependent cell cytotoxicity (ADCC) of primary human eosinophils and basophils and replicated the in vivo depletion capacity of their murine parent. Therefore, depletion of hCRTh2+ basophils, eosinophils, ILC2, and Th2 cells with h19A2 hCRTh2-specific antibodies may be a novel and more efficacious treatment for asthma.


Science Signaling | 2015

Inhibition of the kinase ITK in a mouse model of asthma reduces cell death and fails to inhibit the inflammatory response

Yonglian Sun; Ivan Peng; Joshua D. Webster; Eric Suto; Justin Lesch; Xiumin Wu; Kate Senger; George Francis; Kathy Barrett; Jenna L. Collier; Jason D. Burch; Meijuan Zhou; Yuan Chen; Connie Chan; Jeff Eastham-Anderson; Hai Ngu; Olga Li; Tracy Staton; Charles Havnar; Allan Jaochico; Janet Jackman; Surinder Jeet; Lorena Riol-Blanco; Lawren C. Wu; David F. Choy; Joseph R. Arron; Brent S. McKenzie; Nico Ghilardi; Moulay Hicham Alaoui Ismaili; Zhonghua Pei

The kinase ITK is required for antigen-stimulated T cell death. Targeting ITK in asthma CD4+ T helper 2 (TH2) lymphocytes secrete the cytokines interleukin-4 (IL-4), IL-15, and IL-13, which are implicated in the pathogenesis of asthma. Antigen stimulation of T cells activates the kinase ITK, which is required for TH2-type cytokine production. ITK knockout mice are resistant to airway inflammation, which suggests that ITK inhibitors might be used to treat human asthma. However, Sun et al. found that a mouse model of asthma developed worse disease when treated with an ITK-specific inhibitor, exhibiting increased numbers of T cells and amounts of TH2-type cytokines in the airways. These effects were associated with a failure of ITK-inhibited T cells to undergo antigen-stimulated cell death. Together, these data suggest that targeting the kinase activity of ITK in human asthma may exacerbate disease. Interleukin-2 (IL-2)–inducible T cell kinase (ITK) mediates T cell receptor (TCR) signaling primarily to stimulate the production of cytokines, such as IL-4, IL-5, and IL-13, from T helper 2 (TH2) cells. Compared to wild-type mice, ITK knockout mice are resistant to asthma and exhibit reduced lung inflammation and decreased amounts of TH2-type cytokines in the bronchoalveolar lavage fluid. We found that a small-molecule selective inhibitor of ITK blocked TCR-mediated signaling in cultured TH2 cells, including the tyrosine phosphorylation of phospholipase C–γ1 (PLC-γ1) and the secretion of IL-2 and TH2-type cytokines. Unexpectedly, inhibition of the kinase activity of ITK during or after antigen rechallenge in an ovalbumin-induced mouse model of asthma failed to reduce airway hyperresponsiveness and inflammation. Rather, in mice, pharmacological inhibition of ITK resulted in T cell hyperplasia and the increased production of TH2-type cytokines. Thus, our studies predict that inhibition of the kinase activity of ITK may not be therapeutic in patients with asthma.


PLOS ONE | 2017

Combined MEK and ERK inhibition overcomes therapy-mediated pathway reactivation in RAS mutant tumors

Mark Merchant; John Moffat; Gabriele Schaefer; Jocelyn Chan; Xi Wang; Christine Orr; Jason H. Cheng; Thomas Hunsaker; Lily Shao; Stephanie J. Wang; Marie-Claire Wagle; Eva Lin; Peter M. Haverty; Sheerin Shahidi-Latham; Hai Ngu; Margaret Solon; Jeffrey Eastham-Anderson; Hartmut Koeppen; Shih-Min A. Huang; Jacob B. Schwarz; Marcia Belvin; Daniel C. Kirouac; Melissa R. Junttila

Mitogen-activated protein kinase (MAPK) pathway dysregulation is implicated in >30% of all cancers, rationalizing the development of RAF, MEK and ERK inhibitors. While BRAF and MEK inhibitors improve BRAF mutant melanoma patient outcomes, these inhibitors had limited success in other MAPK dysregulated tumors, with insufficient pathway suppression and likely pathway reactivation. In this study we show that inhibition of either MEK or ERK alone only transiently inhibits the MAPK pathway due to feedback reactivation. Simultaneous targeting of both MEK and ERK nodes results in deeper and more durable suppression of MAPK signaling that is not achievable with any dose of single agent, in tumors where feedback reactivation occurs. Strikingly, combined MEK and ERK inhibition is synergistic in RAS mutant models but only additive in BRAF mutant models where the RAF complex is dissociated from RAS and thus feedback productivity is disabled. We discovered that pathway reactivation in RAS mutant models occurs at the level of CRAF with combination treatment resulting in a markedly more active pool of CRAF. However, distinct from single node targeting, combining MEK and ERK inhibitor treatment effectively blocks the downstream signaling as assessed by transcriptional signatures and phospho-p90RSK. Importantly, these data reveal that MAPK pathway inhibitors whose activity is attenuated due to feedback reactivation can be rescued with sufficient inhibition by using a combination of MEK and ERK inhibitors. The MEK and ERK combination significantly suppresses MAPK pathway output and tumor growth in vivo to a greater extent than the maximum tolerated doses of single agents, and results in improved anti-tumor activity in multiple xenografts as well as in two Kras mutant genetically engineered mouse (GEM) models. Collectively, these data demonstrate that combined MEK and ERK inhibition is functionally unique, yielding greater than additive anti-tumor effects and elucidates a highly effective combination strategy in MAPK-dependent cancer, such as KRAS mutant tumors.


Scientific Reports | 2017

BACE1 across species: a comparison of the in vivo consequences of BACE1 deletion in mice and rats

Martin Weber; Tiffany Wu; William J. Meilandt; Sara L. Dominguez; Hilda Solanoy; Janice Maloney; Hai Ngu; Miriam Baca; Chung Kung; Lisa Lima; Timothy K. Earr; Daniel Fleck; Shannon D. Shields; William F. Forrest; Oded Foreman; Søren Warming; Ryan J. Watts; Kimberly Scearce-Levie

Assessing BACE1 (β-site APP cleaving enzyme 1) knockout mice for general health and neurological function may be useful in predicting risks associated with prolonged pharmacological BACE1 inhibition, a treatment approach currently being developed for Alzheimer’s disease. To determine whether BACE1 deletion-associated effects in mice generalize to another species, we developed a novel Bace1−/− rat line using zinc-finger nuclease technology and compared Bace1−/− mice and rats with their Bace1+/+ counterparts. Lack of BACE1 was confirmed in Bace1−/− animals from both species. Removal of BACE1 affected startle magnitude, balance beam performance, pain response, and nerve myelination in both species. While both mice and rats lacking BACE1 have shown increased mortality, the increase was smaller and restricted to early developmental stages for rats. Bace1−/− mice and rats further differed in body weight, spontaneous locomotor activity, and prepulse inhibition of startle. While the effects of species and genetic background on these phenotypes remain difficult to distinguish, our findings suggest that BACE1’s role in myelination and some sensorimotor functions is consistent between mice and rats and may be conserved in other species. Other phenotypes differ between these models, suggesting that some effects of BACE1 inhibition vary with the biological context (e.g. species or background strain).


Scientific Reports | 2018

Antibody-mediated stabilization of NRG1 induces behavioral and electrophysiological alterations in adult mice

Sara L. Dominguez; Ganapati V. Hegde; Jesse E. Hanson; Hong Xiang; Danielle Mandikian; C. Andrew Boswell; Cecilia Chiu; Yan Wu; Siao Ping Tsai; Daniel Fleck; Martin Weber; Hai Ngu; Kimberly Scearce-Levie; Erica Jackson

Neuregulin 1 (NRG1) is required for development of the central and peripheral nervous system and regulates neurotransmission in the adult. NRG1 and the gene encoding its receptor, ERBB4, are risk genes for schizophrenia, although how alterations in these genes disrupt their function has not been fully established. Studies of knockout and transgenic mice have yielded conflicting results, with both gain and loss of function resulting in similar behavioral and electrophysiological phenotypes. Here, we used high affinity antibodies to NRG1 and ErbB4 to perturb the function of the endogenous proteins in adult mice. Treatment with NRG1 antibodies that block receptor binding caused behavioral alterations associated with schizophrenia, including, hyper-locomotion and impaired pre-pulse inhibition of startle (PPI). Electrophysiological analysis of brain slices from anti-NRG1 treated mice revealed reduced synaptic transmission and enhanced paired-pulse facilitation. In contrast, mice treated with more potent ErbB4 function blocking antibodies did not display behavioral alterations, suggesting a receptor independent mechanism of the anti-NRG1-induced phenotypes. We demonstrate that anti-NRG1 causes accumulation of the full-length transmembrane protein and increases phospho-cofilin levels, which has previously been linked to impaired synaptic transmission, indicating enhancement of non-canonical NRG1 signaling could mediate the CNS effects.

Collaboration


Dive into the Hai Ngu's collaboration.

Researchain Logo
Decentralizing Knowledge