Publications

2024

Huang, Hou-Yuan, Shelbi Salinas, Jessica Cornell, Iquo-Bella Udoh, Yang Shen, and Miou Zhou. (2024) 2024. “CCR5 Regulates Aβ1-42-Induced Learning and Memory Deficits in Mice”. Neurobiology of Learning and Memory 208: 107890. https://doi.org/10.1016/j.nlm.2024.107890.

C-C chemokine receptor 5 (CCR5) is a chemokine receptor involved in immune responses and a co-receptor for HIV infection. Recently, CCR5 has also been reported to play a role in synaptic plasticity, learning and memory, and cognitive deficits associated with normal aging, traumatic brain injury (TBI), and HIV-associated neurocognitive disorder (HAND). In contrast, the role of CCR5 in cognitive deficits associated with other disorders, including Alzheimer's disease (AD), is much less understood. Studies have reported an increase in expression of CCR5 or its ligands in both AD patients and AD rodent models, suggesting a correlation between AD and CCR5 expression. However, whether blocking CCR5 in specific brain regions, such as the hippocampus, could improve memory deficits in AD mouse models is unknown. To study the potential causal role of CCR5 in cognitive deficits in AD, we injected soluble Aβ1-42 or a control (Aβ42-1) oligomers in the dorsal CA1 region of the hippocampus and found that Aβ1-42 injection resulted in severe memory impairment in the object place recognition (OPR) and novel object recognition (NOR) tests. Aβ1-42 injection caused an increase in Ccr5, Ccl3, and Ccl4 in the dorsal hippocampus, and the expression levels of CCR5 and its ligands remained elevated at 2 weeks after Aβ1-42 injection. Knocking down Ccr5 in the CA1 region of dorsal hippocampus reversed the increase in microglia number and size in dorsal CA1 and rescued memory deficits. These results indicate that CCR5 plays an important role in modulating Aβ1-42-induced learning and memory deficits, and suggest that CCR5 antagonists may serve as a potential treatment to improve cognitive deficits associated with AD.

2023

Berard, Lauren, and Miou Zhou. (2023) 2023. “The Impact of Edentulism and Periodontitis on Cognition”. Journal of the California Dental Association 52. https://doi.org/10.1080/19424396.2023.2289696.

Background

Masticatory dysfunction and periodontitis are independently linked to cognitive decline. This article aims to highlight the individual and common roles that periodontitis and tooth loss play in cognition. Specifically, we aim to summarize the behavioral effects on learning and memory that result from each pathology, explore mechanisms that may explain these effects, and review the commonalities between the two.

Methods

The PubMed database was used to implement searches of relevant clinical studies on the correlations between edentulism and cognitive decline as well as periodontitis and cognitive decline. Additional searches were done to investigate animal studies that provided behavioral and mechanistic findings to explain these relationships. The clinical and animal studies were summarized and analyzed in this review to reveal the commonalities and differences between these comorbidities.

Results

The clinical studies summarized in this review report significant correlations between both periodontitis and partial/full edentulism with cognitive decline. The behavioral and mechanistic findings in animal model studies are summarized to support the cognitive effects. Both in vivo and in vitro studies have identified key pathogens, molecules, or cellular pathways that contribute to a more comprehensive understanding of the pathophysiologic processes that link different types of oral disease to cognitive brain function.

Practical Implications

The correlation between oral disease and cognitive impairment has been well established but further exploration of the molecular and cellular mechanisms underlying these relationships can provide therapeutic implications and stress the importance of oral health and preventative care.

2022

Cornell, Jessica, Shelbi Salinas, Hou-Yuan Huang, and Miou Zhou. (2022) 2022. “Microglia Regulation of Synaptic Plasticity and Learning and Memory”. Neural Regeneration Research 17 (4): 705-16. https://doi.org/10.4103/1673-5374.322423.

Microglia are the resident macrophages of the central nervous system. Microglia possess varied morphologies and functions. Under normal physiological conditions, microglia mainly exist in a resting state and constantly monitor their microenvironment and survey neuronal and synaptic activity. Through the C1q, C3 and CR3 "Eat Me" and CD47 and SIRPα "Don't Eat Me" complement pathways, as well as other pathways such as CX3CR1 signaling, resting microglia regulate synaptic pruning, a process crucial for the promotion of synapse formation and the regulation of neuronal activity and synaptic plasticity. By mediating synaptic pruning, resting microglia play an important role in the regulation of experience-dependent plasticity in the barrel cortex and visual cortex after whisker removal or monocular deprivation, and also in the regulation of learning and memory, including the modulation of memory strength, forgetfulness, and memory quality. As a response to brain injury, infection or neuroinflammation, microglia become activated and increase in number. Activated microglia change to an amoeboid shape, migrate to sites of inflammation and secrete proteins such as cytokines, chemokines and reactive oxygen species. These molecules released by microglia can lead to synaptic plasticity and learning and memory deficits associated with aging, Alzheimer's disease, traumatic brain injury, HIV-associated neurocognitive disorder, and other neurological or mental disorders such as autism, depression and post-traumatic stress disorder. With a focus mainly on recently published literature, here we reviewed the studies investigating the role of resting microglia in synaptic plasticity and learning and memory, as well as how activated microglia modulate disease-related plasticity and learning and memory deficits. By summarizing the function of microglia in these processes, we aim to provide an overview of microglia regulation of synaptic plasticity and learning and memory, and to discuss the possibility of microglia manipulation as a therapeutic to ameliorate cognitive deficits associated with aging, Alzheimer's disease, traumatic brain injury, HIV-associated neurocognitive disorder, and mental disorders.

Riviere-Cazaux, Cecile, Jessica Cornell, Yang Shen, and Miou Zhou. (2022) 2022. “The Role of CCR5 in HIV-Associated Neurocognitive Disorders”. Heliyon 8 (7): e09950. https://doi.org/10.1016/j.heliyon.2022.e09950.

While combination antiretroviral therapy (cART) has successfully increased the lifespan of individuals infected with HIV, a significant portion of this population remains affected by HIV-associated neurocognitive disorder (HAND). C-C chemokine receptor 5 (CCR5) has been well studied in immune response and as a co-receptor for HIV infection. HIV-infected (HIV+) patients experienced mild to significant amelioration of cognitive function when treated with different CCR5 antagonists, including maraviroc and cenicriviroc. Consistent with clinical results, Ccr5 knockout or knockdown rescued cognitive deficits in HIV animal models, with mechanisms of reduced microgliosis and neuroinflammation. Pharmacologic inhibition of CCR5 directly improved cerebral and hippocampal neuronal plasticity and cognitive function. By summarizing the animal and human studies of CCR5 in HIV-associated cognitive deficits, this review aims to provide an overview of the mechanistic role of CCR5 in HAND pathophysiology. This review also discusses the addition of CCR5 antagonists, such as maraviroc, to cART for targeted prevention and treatment of cognitive impairments in patients infected with HIV.

Guo, Changyong, Di Wen, Yihong Zhang, Richie Mustaklem, Basil Mustaklem, Miou Zhou, Tao Ma, and Yao-Ying Ma. (2022) 2022. “Amyloid-β Oligomers in the Nucleus Accumbens Decrease Motivation via Insertion of Calcium-Permeable AMPA Receptors”. Molecular Psychiatry 27 (4): 2146-57. https://doi.org/10.1038/s41380-022-01459-0.

It is essential to identify the neuronal mechanisms of Alzheimer's Disease (AD)-associated neuropsychiatric symptoms, e.g., apathy, before improving the life quality of AD patients. Here, we focused on the nucleus accumbens (NAc), a critical brain region processing motivation, also known to display AD-associated pathological changes in human cases. We found that the synaptic calcium permeable (CP)-AMPA receptors (AMPARs), which are normally absent in the NAc, can be revealed by acute exposure to Aβ oligomers (AβOs), and play a critical role in the emergence of synaptic loss and motivation deficits. Blockade of NAc CP-AMPARs can effectively prevent AβO-induced downsizing and pruning of spines and silencing of excitatory synaptic transmission. We conclude that AβO-triggered synaptic insertion of CP-AMPARs is a key mechanism mediating synaptic degeneration in AD, and preserving synaptic integrity may prevent or delay the onset of AD-associated psychiatric symptoms.

Shen, Yang, Miou Zhou, Denise Cai, Daniel Almeida Filho, Giselle Fernandes, Ying Cai, André F de Sousa, et al. (2022) 2022. “CCR5 Closes the Temporal Window for Memory Linking”. Nature 606 (7912): 146-52. https://doi.org/10.1038/s41586-022-04783-1.

Real-world memories are formed in a particular context and are often not acquired or recalled in isolation1-5. Time is a key variable in the organization of memories, as events that are experienced close in time are more likely to be meaningfully associated, whereas those that are experienced with a longer interval are not1-4. How the brain segregates events that are temporally distinct is unclear. Here we show that a delayed (12-24 h) increase in the expression of C-C chemokine receptor type 5 (CCR5)-an immune receptor that is well known as a co-receptor for HIV infection6,7-after the formation of a contextual memory determines the duration of the temporal window for associating or linking that memory with subsequent memories. This delayed expression of CCR5 in mouse dorsal CA1 neurons results in a decrease in neuronal excitability, which in turn negatively regulates neuronal memory allocation, thus reducing the overlap between dorsal CA1 memory ensembles. Lowering this overlap affects the ability of one memory to trigger the recall of the other, and therefore closes the temporal window for memory linking. Our findings also show that an age-related increase in the neuronal expression of CCR5 and its ligand CCL5 leads to impairments in memory linking in aged mice, which could be reversed with a Ccr5 knockout and a drug approved by the US Food and Drug Administration (FDA) that inhibits this receptor, a result with clinical implications. Altogether, the findings reported here provide insights into the molecular and cellular mechanisms that shape the temporal window for memory linking.

2021

Ozkan, Alper D, Tina Gettas, Audrey Sogata, Wynn Phaychanpheng, Miou Zhou, and Jérôme J Lacroix. (2021) 2021. “Mechanical and Chemical Activation of GPR68 Probed With a Genetically Encoded Fluorescent Reporter”. Journal of Cell Science 134 (16). https://doi.org/10.1242/jcs.255455.

G-protein-coupled receptor (GPCR) 68 (GPR68, or OGR1) couples extracellular acidifications and mechanical stimuli to G-protein signaling and plays important roles in vascular physiology, neuroplasticity and cancer progression. Inspired by previous GPCR-based reporters, here, we inserted a cyclic permuted fluorescent protein into the third intracellular loop of GPR68 to create a genetically encoded fluorescent reporter of GPR68 activation we call 'iGlow'. iGlow responds to known physiological GPR68 activators such as fluid shear stress and extracellular acidifications. In addition, iGlow responds to Ogerin, a synthetic GPR68-selective agonist, but not to a non-active Ogerin analog, showing the specificity of iGlow-mediated fluorescence signals. Flow-induced iGlow activation is not eliminated by pharmacological modulation of downstream G-protein signaling, disruption of actin filaments or application of GsMTx4, an inhibitor of certain mechanosensitive ion channels activated by membrane stretch. Deletion of the conserved helix 8, proposed to mediate mechanosensitivity in certain GPCRs, does not eliminate flow-induced iGlow activation. iGlow could be useful to investigate the contribution of GPR68-dependent signaling in health and disease.

López-Aranda, Manuel F, Ishanu Chattopadhyay, Gayle M Boxx, Elizabeth R Fraley, Tawnie K Silva, Miou Zhou, Miranda Phan, et al. (2021) 2021. “Postnatal Immune Activation Causes Social Deficits in a Mouse Model of Tuberous Sclerosis: Role of Microglia and Clinical Implications”. Science Advances 7 (38): eabf2073. https://doi.org/10.1126/sciadv.abf2073.

There is growing evidence that prenatal immune activation contributes to neuropsychiatric disorders. Here, we show that early postnatal immune activation resulted in profound impairments in social behavior, including in social memory in adult male mice heterozygous for a gene responsible for tuberous sclerosis complex (Tsc2+/−), a genetic disorder with high prevalence of autism. Early postnatal immune activation did not affect either wild-type or female Tsc2+/− mice. We demonstrate that these memory deficits are caused by abnormal mammalian target of rapamycin–dependent interferon signaling and impairments in microglia function. By mining the medical records of more than 3 million children followed from birth, we show that the prevalence of hospitalizations due to infections in males (but not in females) is associated with future development of autism spectrum disorders (ASD). Together, our results suggest the importance of synergistic interactions between strong early postnatal immune activation and mutations associated with ASD.

Necula, Deanna, Cecile Riviere-Cazaux, Yang Shen, and Miou Zhou. (2021) 2021. “Insight into the Roles of CCR5 in Learning and Memory in Normal and Disordered States”. Brain, Behavior, and Immunity 92: 1-9. https://doi.org/10.1016/j.bbi.2020.11.037.

As cognitive impairments continue to rise in prevalence, there is an urgent need to understand the mechanisms of learning and memory in normal and disordered states. C-C chemokine receptor 5 (CCR5) has been implicated in the regulation of multiple forms of learning and memory via its regulation on learning-related cell signaling and neuronal plasticity. As a chemokine receptor and a co-receptor for HIV, CCR5's role in immune response and HIV-associated neurocognitive disorder (HAND) has been widely studied. In contrast, CCR5 is less understood in cognitive deficits associated with other disorders, including Alzheimer's disease (AD), stroke and certain psychiatric disorders. A broad overview of the present literature shows that CCR5 acts as a potent suppressor of synaptic plasticity and learning and memory, although a few studies have reported the opposite effect of CCR5 in stroke or AD animal models. By summarizing the current literature of CCR5 in animal and human studies of cognition, this review aims to provide a comprehensive overview of the role of CCR5 in learning and memory in both normal and disordered states and to discuss the possibility of CCR5 suppression as an effective therapeutic to alleviate cognitive deficits in HAND, AD, and stroke.

2019

Joy, Mary T, Einor Ben Assayag, Dalia Shabashov-Stone, Sigal Liraz-Zaltsman, Jose Mazzitelli, Marcela Arenas, Nora Abduljawad, et al. (2019) 2019. “CCR5 Is a Therapeutic Target for Recovery After Stroke and Traumatic Brain Injury”. Cell 176 (5): 1143-1157.e13. https://doi.org/10.1016/j.cell.2019.01.044.

We tested a newly described molecular memory system, CCR5 signaling, for its role in recovery after stroke and traumatic brain injury (TBI). CCR5 is uniquely expressed in cortical neurons after stroke. Post-stroke neuronal knockdown of CCR5 in pre-motor cortex leads to early recovery of motor control. Recovery is associated with preservation of dendritic spines, new patterns of cortical projections to contralateral pre-motor cortex, and upregulation of CREB and DLK signaling. Administration of a clinically utilized FDA-approved CCR5 antagonist, devised for HIV treatment, produces similar effects on motor recovery post stroke and cognitive decline post TBI. Finally, in a large clinical cohort of stroke patients, carriers for a naturally occurring loss-of-function mutation in CCR5 (CCR5-Δ32) exhibited greater recovery of neurological impairments and cognitive function. In summary, CCR5 is a translational target for neural repair in stroke and TBI and the first reported gene associated with enhanced recovery in human stroke.