Network


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

Hotspot


Dive into the research topics where Alex G. Lee is active.

Publication


Featured researches published by Alex G. Lee.


Nature | 2002

Prokineticin 2 transmits the behavioural circadian rhythm of the suprachiasmatic nucleus

Michelle Y. Cheng; Clayton M. Bullock; Chuanyu Li; Alex G. Lee; Jason C. Bermak; James D. Belluzzi; David R. Weaver; Frances M. Leslie; Qun-Yong Zhou

The suprachiasmatic nucleus (SCN) controls the circadian rhythm of physiological and behavioural processes in mammals. Here we show that prokineticin 2 (PK2), a cysteine-rich secreted protein, functions as an output molecule from the SCN circadian clock. PK2 messenger RNA is rhythmically expressed in the SCN, and the phase of PK2 rhythm is responsive to light entrainment. Molecular and genetic studies have revealed that PK2 is a gene that is controlled by a circadian clock (clock-controlled). Receptor for PK2 (PKR2) is abundantly expressed in major target nuclei of the SCN output pathway. Inhibition of nocturnal locomotor activity in rats by intracerebroventricular delivery of recombinant PK2 during subjective night, when the endogenous PK2 mRNA level is low, further supports the hypothesis that PK2 is an output molecule that transmits behavioural circadian rhythm. The high expression of PKR2 mRNA within the SCN and the positive feedback of PK2 on its own transcription through activation of PKR2 suggest that PK2 may also function locally within the SCN to synchronize output.


The Journal of Neuroscience | 2006

Attenuated circadian rhythms in mice lacking the prokineticin 2 gene.

Jia-Da Li; Wang-Ping Hu; Lisa Boehmer; Michelle Y. Cheng; Alex G. Lee; Alexander Jilek; Jerome M. Siegel; Qun-Yong Zhou

Circadian clocks drive daily rhythms in virtually all organisms. In mammals, the suprachiasmatic nucleus (SCN) is recognized as the master clock that synchronizes central and peripheral oscillators to evoke circadian rhythms of diverse physiology and behavior. How the timing information is transmitted from the SCN clock to generate overt circadian rhythms is essentially unknown. Prokineticin 2 (PK2), a clock-controlled gene that encodes a secreted protein, has been indicated as a candidate SCN clock output signal that regulates circadian locomotor rhythm. Here we report the generation and analysis of PK2-null mice. The reduction of locomotor rhythms in PK2-null mice was apparent in both hybrid and inbred genetic backgrounds. PK2-null mice also displayed significantly reduced rhythmicity for a variety of other physiological and behavioral parameters, including sleep–wake cycle, body temperature, circulating glucocorticoid and glucose levels, as well as the expression of peripheral clock genes. In addition, PK2-null mice showed accelerated acquisition of food anticipatory activity during a daytime food restriction. We conclude that PK2, acting as a SCN output factor, is important for the maintenance of robust circadian rhythms.


Nature Medicine | 2014

Epigenetic targeting of Hedgehog pathway transcriptional output through BET bromodomain inhibition

Yujie Tang; Sharareh Gholamin; Simone Schubert; Minde Willardson; Alex G. Lee; Pratiti Bandopadhayay; Guillame Bergthold; Sabran Masoud; Brian Nguyen; Nujsaubnusi Vue; Brianna Balansay; Furong Yu; Sekyung Oh; Pamelyn Woo; Spenser Chen; Anitha Ponnuswami; Michelle Monje; Scott X. Atwood; Ramon J. Whitson; Siddhartha Mitra; Samuel H. Cheshier; Jun Qi; Rameen Beroukhim; Jean Y. Tang; Rob Wechsler-Reya; Anthony E. Oro; Brian A. Link; James E. Bradner; Yoon-Jae Cho

Hedgehog signaling drives oncogenesis in several cancers, and strategies targeting this pathway have been developed, most notably through inhibition of Smoothened (SMO). However, resistance to Smoothened inhibitors occurs by genetic changes of Smoothened or other downstream Hedgehog components. Here we overcome these resistance mechanisms by modulating GLI transcription through inhibition of bromo and extra C-terminal (BET) bromodomain proteins. We show that BRD4 and other BET bromodomain proteins regulate GLI transcription downstream of SMO and suppressor of fused (SUFU), and chromatin immunoprecipitation studies reveal that BRD4 directly occupies GLI1 and GLI2 promoters, with a substantial decrease in engagement of these sites after treatment with JQ1, a small-molecule inhibitor targeting BRD4. Globally, genes associated with medulloblastoma-specific GLI1 binding sites are downregulated in response to JQ1 treatment, supporting direct regulation of GLI activity by BRD4. Notably, patient- and GEMM (genetically engineered mouse model)-derived Hedgehog-driven tumors (basal cell carcinoma, medulloblastoma and atypical teratoid rhabdoid tumor) respond to JQ1 even when harboring genetic lesions rendering them resistant to Smoothened antagonists. Altogether, our results reveal BET proteins as critical regulators of Hedgehog pathway transcriptional output and nominate BET bromodomain inhibitors as a strategy for treating Hedgehog-driven tumors with emerged or a priori resistance to Smoothened antagonists.


Nature | 2013

GLI activation by atypical protein kinase C ι/λ regulates the growth of basal cell carcinomas.

Scott X. Atwood; Mischa Li; Alex G. Lee; Jean Y. Tang; Anthony E. Oro

Growth of basal cell carcinomas (BCCs) requires high levels of hedgehog (HH) signalling through the transcription factor GLI. Although inhibitors of membrane protein smoothened (SMO) effectively suppress HH signalling, early tumour resistance illustrates the need for additional downstream targets for therapy. Here we identify atypical protein kinase C ι/λ (aPKC-ι/λ) as a novel GLI regulator in mammals. aPKC-ι/λ and its polarity signalling partners co-localize at the centrosome and form a complex with missing-in-metastasis (MIM), a scaffolding protein that potentiates HH signalling. Genetic or pharmacological loss of aPKC-ι/λ function blocks HH signalling and proliferation of BCC cells. Prkci is a HH target gene that forms a positive feedback loop with GLI and exists at increased levels in BCCs. Genome-wide transcriptional profiling shows that aPKC-ι/λ and SMO control the expression of similar genes in tumour cells. aPKC-ι/λ functions downstream of SMO to phosphorylate and activate GLI1, resulting in maximal DNA binding and transcriptional activation. Activated aPKC-ι/λ is upregulated in SMO-inhibitor-resistant tumours and targeting aPKC-ι/λ suppresses signalling and growth of resistant BCC cell lines. These results demonstrate that aPKC-ι/λ is critical for HH-dependent processes and implicates aPKC-ι/λ as a new, tumour-selective therapeutic target for the treatment of SMO-inhibitor-resistant cancers.Growth of basal cell carcinomas (BCCs) requires high levels of hedgehog (HH) signalling through the transcription factor GLI. Although inhibitors of membrane protein smoothened (SMO) effectively suppress HH signalling, early tumour resistance illustrates the need for additional downstream targets for therapy. Here we identify atypical protein kinase C ι/λ (aPKC-ι/λ) as a novel GLI regulator in mammals. aPKC-ι/λ and its polarity signalling partners co-localize at the centrosome and form a complex with missing-in-metastasis (MIM), a scaffolding protein that potentiates HH signalling. Genetic or pharmacological loss of aPKC-ι/λ function blocks HH signalling and proliferation of BCC cells. Prkci is a HH target gene that forms a positive feedback loop with GLI and exists at increased levels in BCCs. Genome-wide transcriptional profiling shows that aPKC-ι/λ and SMO control the expression of similar genes in tumour cells. aPKC-ι/λ functions downstream of SMO to phosphorylate and activate GLI1, resulting in maximal DNA binding and transcriptional activation. Activated aPKC-ι/λ is upregulated in SMO-inhibitor-resistant tumours and targeting aPKC-ι/λ suppresses signalling and growth of resistant BCC cell lines. These results demonstrate that aPKC-ι/λ is critical for HH-dependent processes and implicates aPKC-ι/λ as a new, tumour-selective therapeutic target for the treatment of SMO-inhibitor-resistant cancers.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Stress coping stimulates hippocampal neurogenesis in adult monkeys

David M. Lyons; Paul S. Buckmaster; Alex G. Lee; Christine Wu; Rupshi Mitra; Lauren M Duffey; Christine L. Buckmaster; Song Her; Paresh D. Patel; Alan F. Schatzberg

Coping with intermittent social stress is an essential aspect of living in complex social environments. Coping tends to counteract the deleterious effects of stress and is thought to induce neuroadaptations in corticolimbic brain systems. Here we test this hypothesis in adult squirrel monkey males exposed to intermittent social separations and new pair formations. These manipulations simulate conditions that typically occur in male social associations because of competition for limited access to residency in mixed-sex groups. As evidence of coping, we previously confirmed that cortisol levels initially increase and then are restored to prestress levels within several days of each separation and new pair formation. Follow-up studies with exogenous cortisol further established that feedback regulation of the hypothalamic-pituitary-adrenal axis is not impaired. Now we report that exposure to intermittent social separations and new pair formations increased hippocampal neurogenesis in squirrel monkey males. Hippocampal neurogenesis in rodents contributes to spatial learning performance, and in monkeys we found that spatial learning was enhanced in conditions that increased hippocampal neurogenesis. Corresponding changes were discerned in the expression of genes involved in survival and integration of adult-born granule cells into hippocampal neural circuits. These findings support recent indications that stress coping stimulates hippocampal neurogenesis in adult rodents. Psychotherapies designed to promote stress coping potentially have similar effects in humans with major depression.


Biology Letters | 2011

A novel form of oxytocin in New World monkeys.

Alex G. Lee; David R. Cool; William C. Grunwald Jr.; Donald E. Neal; Christine L. Buckmaster; Michelle Y. Cheng; Shellie A. Hyde; David M. Lyons; Karen J. Parker

Oxytocin is widely believed to be present and structurally identical in all placental mammals. Here, we report that multiple species of New World monkeys possess a novel form of oxytocin, [P8] oxytocin. This mutation arises from a substitution of a leucine to a proline in amino acid position 8. Further analysis of this mutation in Saimiri sciureus (squirrel monkey) indicates that [P8] oxytocin is transcribed and translated properly. This mutation is specific to oxytocin, as the peptide sequence for arginine vasopressin, a structurally related nonapeptide, is unaltered. These findings dispel the notion that all placental mammals possess a ‘universal’ oxytocin sequence, and highlight the need for research on the functional significance of this novel nonapeptide in New World monkeys.


Journal of Non-newtonian Fluid Mechanics | 2002

A study of viscoelastic free surface flows by the finite element method: Hele–Shaw and slot coating flows

Alex G. Lee; Eric S. G. Shaqfeh; Bamin Khomami

Abstract A pseudo-solid domain mapping technique coupled with the DEVSS finite element formulation is applied to study the effects of viscoelasticity on free surface flows. Two distinct flow types are analyzed: the flow induced by a long air bubble steadily displacing a polymeric liquid confined by two parallel plates, i.e. Hele–Shaw flow, and the slot coating of viscoelastic fluids in the low metering rate limit. The Oldroyd-B, FENE-CR, and FENE-P constitutive equations are used to model the viscoelastic fluid. Our study reveals the formation of an elastic boundary layer in the capillary-transition region near the bubble front at moderate Weissenberg numbers while the stress field in the parallel flow region remains largely unaffected by the dynamics of the free surface. Our calculations show that the increase in the hydrodynamic coating thickness due to viscoelasticity often reported in planar injection experiments [Physica A 220 (1995) 60; J. Non-Newtonian Fluid Mech. 71 (1997) 73] is associated with the onset of these elastic boundary layers and is strongly dependent on the physical properties of the coating fluid.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Optogenetic neuronal stimulation promotes functional recovery after stroke.

Michelle Y. Cheng; Eric H Wang; Wyatt J. Woodson; Stephanie Wang; Guohua Sun; Alex G. Lee; Ahmet Arac; Lief E. Fenno; Karl Deisseroth; Gary K. Steinberg

Significance Stroke is the leading cause of disability in the United States and has very limited treatment options. Brain stimulation techniques that promote recovery after stroke are a promising area of research; however, current stimulation techniques nonspecifically activate/inhibit the target area, which not only leads to undesired side effects but also makes it difficult to understand which cell types and mechanisms drive recovery. We used the optogenetic technique to specifically stimulate only neurons after stroke and demonstrate that selective neuronal stimulations can activate beneficial mechanisms and promote recovery. Understanding the cell type and mechanisms driving recovery may identify potential drug targets for stroke treatment, as well as ultimately help develop precise brain stimulation techniques for stroke therapy. Clinical and research efforts have focused on promoting functional recovery after stroke. Brain stimulation strategies are particularly promising because they allow direct manipulation of the target area’s excitability. However, elucidating the cell type and mechanisms mediating recovery has been difficult because existing stimulation techniques nonspecifically target all cell types near the stimulated site. To circumvent these barriers, we used optogenetics to selectively activate neurons that express channelrhodopsin 2 and demonstrated that selective neuronal stimulations in the ipsilesional primary motor cortex (iM1) can promote functional recovery. Stroke mice that received repeated neuronal stimulations exhibited significant improvement in cerebral blood flow and the neurovascular coupling response, as well as increased expression of activity-dependent neurotrophins in the contralesional cortex, including brain-derived neurotrophic factor, nerve growth factor, and neurotrophin 3. Western analysis also indicated that stimulated mice exhibited a significant increase in the expression of a plasticity marker growth-associated protein 43. Moreover, iM1 neuronal stimulations promoted functional recovery, as stimulated stroke mice showed faster weight gain and performed significantly better in sensory-motor behavior tests. Interestingly, stimulations in normal nonstroke mice did not alter motor behavior or neurotrophin expression, suggesting that the prorecovery effect of selective neuronal stimulations is dependent on the poststroke environment. These results demonstrate that stimulation of neurons in the stroke hemisphere is sufficient to promote recovery.


The Journal of Comparative Neurology | 2006

Nicotine modulation of stress‐related peptide neurons

Sandra E. Loughlin; Michelle I. Islas; Michelle Y. Cheng; Alex G. Lee; Anne-Sophie Villégier; Frances M. Leslie

Nicotine has been shown to activate stress‐related brain nuclei, including the paraventricular nucleus of the hypothalamus (PVN) and the central nucleus of the amygdala (CEA), through complex mechanisms involving direct and indirect pathways. To determine the neurochemical identities of rat brain neurons which are activated by a low dose (0.175 mg/kg) of nicotine given 30 minutes before sacrifice, we have used single‐ and double‐label in situ hybridization. Neuronal activation was quantified by localization of 35S‐labeled probe for the immediate early gene, c‐fos. Corticotrophin releasing factor (CRF), enkephalin (ENK), and dynorphin (DYN) mRNAs were colocalized using a colorimetric, digoxigenin‐labeled probe. Film autoradiographic studies showed that nicotine significantly increased c‐fos mRNA expression in both PVN and CEA. Pretreatment with the centrally acting nicotinic antagonist, mecamylamine (1 mg/kg), blocked nicotines effects, whereas pretreatment with the peripherally acting antagonist, hexamethonium (5 mg/kg), did not, indicating that c‐fos induction was mediated by a central nicotinic receptor. Double labeling studies showed that nicotine induced c‐fos expression within CRF cells in the PVN, as well as in a small population of ENK cells, but not in PVN DYN cells. In contrast, there was no significant nicotine‐induced increase in c‐fos expression in CEA CRF or DYN cells, whereas nicotine treatment did increase c‐fos expression within CEA ENK cells. J. Comp. Neurol. 497:575–588, 2006.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Prokineticin 2 is an endangering mediator of cerebral ischemic injury

Michelle Y. Cheng; Alex G. Lee; Collin J. Culbertson; Guohua Sun; Rushi K. Talati; Nathan C. Manley; Xiaohan Li; Heng Zhao; David M. Lyons; Qun-Yong Zhou; Gary K. Steinberg; Robert M. Sapolsky

Stroke causes brain dysfunction and neuron death, and the lack of effective therapies heightens the need for new therapeutic targets. Here we identify prokineticin 2 (PK2) as a mediator for cerebral ischemic injury. PK2 is a bioactive peptide initially discovered as a regulator of gastrointestinal motility. Multiple biological roles for PK2 have been discovered, including circadian rhythms, angiogenesis, and neurogenesis. However, the role of PK2 in neuropathology is unknown. Using primary cortical cultures, we found that PK2 mRNA is up-regulated by several pathological stressors, including hypoxia, reactive oxygen species, and excitotoxic glutamate. Glutamate-induced PK2 expression is dependent on NMDA receptor activation and extracellular calcium. Enriched neuronal culture studies revealed that neurons are the principal source of glutamate-induced PK2. Using in vivo models of stroke, we found that PK2 mRNA is induced in the ischemic cortex and striatum. Central delivery of PK2 worsens infarct volume, whereas PK2 receptor antagonist decreases infarct volume and central inflammation while improving functional outcome. Direct central inhibition of PK2 using RNAi also reduces infarct volume. These findings indicate that PK2 can be activated by pathological stimuli such as hypoxia-ischemia and excitotoxic glutamate and identify PK2 as a deleterious mediator for cerebral ischemia.

Collaboration


Dive into the Alex G. Lee's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Qun-Yong Zhou

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge