1
|
Tasevski S, Kyung Nam H, Ghannam A, Moughni S, Atoui T, Mashal Y, Hatch N, Zhang Z. Tissue nonspecific alkaline phosphatase deficiency impairs Purkinje cell development and survival in a mouse model of infantile hypophosphatasia. Neuroscience 2024; 560:357-370. [PMID: 39369942 DOI: 10.1016/j.neuroscience.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 09/26/2024] [Accepted: 10/02/2024] [Indexed: 10/08/2024]
Abstract
Loss-of-function mutations in the tissue-nonspecific alkaline phosphatase (TNAP) gene can result in hypophosphatasia (HPP), an inherited multi-systemic metabolic disorder that is well-known for skeletal and dental hypomineralization. However, emerging evidence shows that both adult and pediatric patients with HPP suffer from cognitive deficits, higher measures of depression and anxiety, and impaired sensorimotor skills. The cerebellum plays an important role in sensorimotor coordination, cognition, and emotion. To date, the impact of TNAP mutation on the cerebellar circuitry development and function remains poorly understood. The main objective of this study was to investigate the roles of TNAP in cerebellar development and function, with a particular focus on Purkinje cells, in a mouse model of infantile HPP. Male and female wild type (WT) and TNAP knockout (KO) mice underwent behavioral testing on postnatal day 13-14 and were euthanized after completion of behavioral tests. Cerebellar tissues were harvested for gene expression and immunohistochemistry analyses. We found that TNAP mutation resulted in significantly reduced body weight, shorter body length, and impaired sensorimotor functions in both male and female KO mice. These developmental and behavioral deficits were accompanied by abnormal Purkinje cell morphology and dysregulation of genes that regulates synaptic transmission, cellular growth, proliferation, and death. In conclusion, inactivation of TNAP via gene deletion causes developmental delays, sensorimotor impairment, and Purkinje cell maldevelopment. These results shed light on a new perspective of cerebellar dysfunction in HPP.
Collapse
Affiliation(s)
- Stefanie Tasevski
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI 48128, USA
| | - Hwa Kyung Nam
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan-Ann Arbor, 1011 N University Ave, Ann Arbor, MI 48109, USA
| | - Amanda Ghannam
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI 48128, USA
| | - Sara Moughni
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI 48128, USA
| | - Tia Atoui
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI 48128, USA
| | - Yara Mashal
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI 48128, USA
| | - Nan Hatch
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan-Ann Arbor, 1011 N University Ave, Ann Arbor, MI 48109, USA
| | - Zhi Zhang
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI 48128, USA.
| |
Collapse
|
2
|
Wang Y, Qin Y, Zeng S, Zhang Z, Liu W, Wan J, Qian K, Li S, Xiao J. Proteomic and metabolomic analysis of plasma for pain at different labor stages. Talanta 2024; 282:126905. [PMID: 39341059 DOI: 10.1016/j.talanta.2024.126905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 08/29/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024]
Abstract
Labor pain has an important impact on maternal labor experience, mood, and postpartum depression. It is of great emotional significance to pay attention to the pain stress response of pregnant women and take necessary intervention measures in the labor process to weaken the sense of delivery experience and reduce the risk of complications. To better understand the molecular alteration of pain and stress changes during the delivery, we analyzed the metabolomic and proteomic of the plasma collected during the labor process at different stages, revealing the significant changes in metabolites and proteins and the key regulatory pathways. The KEGG enrichment analysis showed the differentially expressed metabolites and differentially expressed proteins were mainly enriched in glutamate metabolism, glutathione metabolism, oxidative phosphorylation, glycolysis/gluconeogenesis, and citrate cycle (TCA cycle). In particular, the glutathione metabolism played a major role in the metabolic pathway of the whole labor process. The result demonstrated the potential significance of the glutathione metabolic pathway in pain regulation.
Collapse
Affiliation(s)
- Yating Wang
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
| | - Yi Qin
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
| | - Shanshan Zeng
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Ziyue Zhang
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Wanshan Liu
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Jingjing Wan
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
| | - Kun Qian
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Shunxiang Li
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, China.
| | - Jie Xiao
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China.
| |
Collapse
|
3
|
Pereira SDC, Manhães-de-Castro R, Souza VDS, Calado CMSDS, Souza de Silveira B, Barbosa LNF, Torner L, Guzmán-Quevedo O, Toscano AE. Neonatal resveratrol treatment in cerebral palsy model recovers neurodevelopment impairments by restoring the skeletal muscle morphology and decreases microglial activation in the cerebellum. Exp Neurol 2024; 378:114835. [PMID: 38789024 DOI: 10.1016/j.expneurol.2024.114835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/11/2024] [Accepted: 05/21/2024] [Indexed: 05/26/2024]
Abstract
Cerebral Palsy (CP) is the main motor disorder in childhood resulting from damage to the developing brain. Treatment perspectives are required to reverse the primary damage caused by the early insult and consequently to recover motor skills. Resveratrol has been shown to act as neuroprotection with benefits to skeletal muscle. This study aimed to investigate the effects of neonatal resveratrol treatment on neurodevelopment, skeletal muscle morphology, and cerebellar damage in CP model. Wistar rat pups were allocated to four experimental groups (n = 15/group) according CP model and treatment: Control+Saline (CS), Control+Resveratrol (CR), CP + Saline (CPS), and CP + Resveratrol (CPR). CP model associated anoxia and sensorimotor restriction. CP group showed delay in the disappearance of the palmar grasp reflex (p < 0.0001) and delay in the appearance of reflexes of negative geotaxis (p = 0.01), and free-fall righting (p < 0.0001), reduced locomotor activity and motor coordination (p < 0.05) than CS group. These motor skills impairments were associated with a reduction in muscle weight (p < 0.001) and area and perimeter of soleus end extensor digitorum longus muscle fibers (p < 0.0001), changes in muscle fibers typing pattern (p < 0.05), and the cerebellum showed signs of neuroinflammation due to elevated density and percentage of activated microglia in the CPS group compared to CS group (p < 0.05). CP animals treated with resveratrol showed anticipation of the appearance of negative geotaxis and free-fall righting reflexes (p < 0.01), increased locomotor activity (p < 0.05), recovery muscle fiber types pattern (p < 0.05), and reversal of the increase in density and the percentage of activated microglia in the cerebellum (p < 0.01). Thus, we conclude that neonatal treatment with resveratrol can contribute to the recovery of the delay neurodevelopment resulting from experimental CP due to its action in restoring the skeletal muscle morphology and reducing neuroinflammation from cerebellum.
Collapse
Affiliation(s)
- Sabrina da Conceição Pereira
- Studies in Nutrition and Phenotypic Plasticity Unit, Center for Health Sciences, Federal University of Pernambuco, Recife, Pernambuco, 50670-420, Brazil; Graduate Program in Neuropsychiatry and Behavioral Sciences, Center for Medical Sciences, Federal University of Pernambuco, Recife, Pernambuco, 50670-901, Brazil
| | - Raul Manhães-de-Castro
- Studies in Nutrition and Phenotypic Plasticity Unit, Center for Health Sciences, Federal University of Pernambuco, Recife, Pernambuco, 50670-420, Brazil; Graduate Program in Neuropsychiatry and Behavioral Sciences, Center for Medical Sciences, Federal University of Pernambuco, Recife, Pernambuco, 50670-901, Brazil; Graduate Program in Nutrition, Center for Health Sciences, Federal University of Pernambuco, Recife, Pernambuco, 50670-420, Brazil
| | - Vanessa da Silva Souza
- Studies in Nutrition and Phenotypic Plasticity Unit, Center for Health Sciences, Federal University of Pernambuco, Recife, Pernambuco, 50670-420, Brazil; Graduate Program in Neuropsychiatry and Behavioral Sciences, Center for Medical Sciences, Federal University of Pernambuco, Recife, Pernambuco, 50670-901, Brazil
| | - Caio Matheus Santos da Silva Calado
- Studies in Nutrition and Phenotypic Plasticity Unit, Center for Health Sciences, Federal University of Pernambuco, Recife, Pernambuco, 50670-420, Brazil; Graduate Program in Neuropsychiatry and Behavioral Sciences, Center for Medical Sciences, Federal University of Pernambuco, Recife, Pernambuco, 50670-901, Brazil
| | - Beatriz Souza de Silveira
- Studies in Nutrition and Phenotypic Plasticity Unit, Center for Health Sciences, Federal University of Pernambuco, Recife, Pernambuco, 50670-420, Brazil; Graduate Program in Neuropsychiatry and Behavioral Sciences, Center for Medical Sciences, Federal University of Pernambuco, Recife, Pernambuco, 50670-901, Brazil
| | - Letícia Nicoly Ferreira Barbosa
- Studies in Nutrition and Phenotypic Plasticity Unit, Center for Health Sciences, Federal University of Pernambuco, Recife, Pernambuco, 50670-420, Brazil
| | - Luz Torner
- Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, 58330, Morelia, Michoacán, Mexico
| | - Omar Guzmán-Quevedo
- Centro Laboratory of Experimental Neuronutrition and Food Engineering, Tecnológico Nacional de México (TECNM), Instituto Tecnológico Superior de Tacámbaro, 61651, Tacámbaro, Michoacán, Mexico
| | - Ana Elisa Toscano
- Studies in Nutrition and Phenotypic Plasticity Unit, Center for Health Sciences, Federal University of Pernambuco, Recife, Pernambuco, 50670-420, Brazil; Graduate Program in Neuropsychiatry and Behavioral Sciences, Center for Medical Sciences, Federal University of Pernambuco, Recife, Pernambuco, 50670-901, Brazil; Graduate Program in Nutrition, Center for Health Sciences, Federal University of Pernambuco, Recife, Pernambuco, 50670-420, Brazil; Nursing Unit, Vitória Academic Center, Federal University of Pernambuco, Vitória de Santo Antão, Pernambuco, 55608-680, Brazil.
| |
Collapse
|
4
|
Ghannam A, Hahn V, Fan J, Tasevski S, Moughni S, Li G, Zhang Z. Sex-specific and cell-specific regulation of ER stress and neuroinflammation after traumatic brain injury in juvenile mice. Exp Neurol 2024; 377:114806. [PMID: 38701941 DOI: 10.1016/j.expneurol.2024.114806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/14/2024] [Accepted: 04/30/2024] [Indexed: 05/06/2024]
Abstract
Endoplasmic reticulum (ER) stress and neuroinflammation play an important role in secondary brain damage after traumatic brain injury (TBI). Due to the complex brain cytoarchitecture, multiple cell types are affected by TBI. However, cell type-specific and sex-specific responses to ER stress and neuroinflammation remain unclear. Here we investigated differential regulation of ER stress and neuroinflammatory pathways in neurons and microglia during the acute phase post-injury in a mouse model of impact acceleration TBI in both males and females. We found that TBI resulted in significant weight loss only in males, and sensorimotor impairment and depressive-like behaviors in both males and females at the acute phase post-injury. By concurrently isolating neurons and microglia from the same brain sample of the same animal, we were able to evaluate the simultaneous responses in neurons and microglia towards ER stress and neuroinflammation in both males and females. We discovered that the ER stress and anti-inflammatory responses were significantly stronger in microglia, especially in female microglia, compared with the male and female neurons. Whereas the degree of phosphorylated-tau (pTau) accumulation was significantly higher in neurons, compared with the microglia. In conclusion, TBI resulted in behavioral deficits and cell type-specific and sex-specific responses to ER stress and neuroinflammation, and abnormal protein accumulation at the acute phase after TBI in immature mice.
Collapse
Affiliation(s)
- Amanda Ghannam
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI 48128, United States of America.
| | - Victoria Hahn
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI 48128, United States of America.
| | - Jie Fan
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI 48128, United States of America.
| | - Stefanie Tasevski
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI 48128, United States of America.
| | - Sara Moughni
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI 48128, United States of America.
| | - Gengxin Li
- Statistics, Department of Mathematics and Statistics, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI 48128, United States of America.
| | - Zhi Zhang
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI 48128, United States of America.
| |
Collapse
|
5
|
Joyce MKP, Uchendu S, Arnsten AFT. Stress and Inflammation Target Dorsolateral Prefrontal Cortex Function: Neural Mechanisms Underlying Weakened Cognitive Control. Biol Psychiatry 2024:S0006-3223(24)01420-3. [PMID: 38944141 DOI: 10.1016/j.biopsych.2024.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 06/15/2024] [Accepted: 06/22/2024] [Indexed: 07/01/2024]
Abstract
Most mental disorders involve dysfunction of the dorsolateral prefrontal cortex (dlPFC), a recently evolved brain region that subserves working memory, abstraction, and the thoughtful regulation of attention, action, and emotion. For example, schizophrenia, depression, long COVID, and Alzheimer's disease are all associated with dlPFC dysfunction, with neuropathology often being focused in layer III. The dlPFC has extensive top-down projections, e.g., to the posterior association cortices to regulate attention and to the subgenual cingulate cortex via the rostral and medial PFC to regulate emotional responses. However, the dlPFC is particularly dependent on arousal state and is very vulnerable to stress and inflammation, which are etiological and/or exacerbating factors for most mental disorders. The cellular mechanisms by which stress and inflammation impact the dlPFC are a topic of current research and are summarized in this review. For example, the layer III dlPFC circuits that generate working memory-related neuronal firing have unusual neurotransmission, depending on NMDA receptor and nicotinic α7 receptor actions that are blocked under inflammatory conditions by kynurenic acid. These circuits also have unusual neuromodulation, with the molecular machinery to magnify calcium signaling in spines needed to support persistent firing, which must be tightly regulated to prevent toxic calcium actions. Stress rapidly weakens layer III connectivity by driving feedforward calcium-cAMP (cyclic adenosine monophosphate) opening of potassium channels on spines. This is regulated by postsynaptic noradrenergic α2A adrenergic receptor and mGluR3 (metabotropic glutamate receptor 3) signaling but dysregulated by inflammation and/or chronic stress exposure, which contribute to spine loss. Treatments that strengthen the dlPFC via pharmacological (the α2A adrenergic receptor agonist, guanfacine) or repetitive transcranial magnetic stimulation manipulation provide a rational basis for therapy.
Collapse
Affiliation(s)
- Mary Kate P Joyce
- Department of Neuroscience, Yale Medical School, New Haven, Connecticut
| | - Stacy Uchendu
- Department of Neuroscience, Yale Medical School, New Haven, Connecticut
| | - Amy F T Arnsten
- Department of Neuroscience, Yale Medical School, New Haven, Connecticut.
| |
Collapse
|
6
|
Su Y, Huang M, Thomas AG, Maragakis J, Huizar KDJ, Zheng Y, Wu Y, Farah MH, Slusher BS. GCPII Inhibition Promotes Remyelination after Peripheral Nerve Injury in Aged Mice. Int J Mol Sci 2024; 25:6893. [PMID: 39000003 PMCID: PMC11241013 DOI: 10.3390/ijms25136893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 06/18/2024] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
Peripheral nerve injuries (PNIs) represent a significant clinical challenge, particularly in elderly populations where axonal remyelination and regeneration are impaired. Developing therapies to enhance these processes is crucial for improving PNI repair outcomes. Glutamate carboxypeptidase II (GCPII) is a neuropeptidase that plays a pivotal role in modulating glutamate signaling through its enzymatic cleavage of the abundant neuropeptide N-acetyl aspartyl glutamate (NAAG) to liberate glutamate. Within the PNS, GCPII is expressed in Schwann cells and activated macrophages, and its expression is amplified with aging. In this study, we explored the therapeutic potential of inhibiting GCPII activity following PNI. We report significant GCPII protein and activity upregulation following PNI, which was normalized by the potent and selective GCPII inhibitor 2-(phosphonomethyl)-pentanedioic acid (2-PMPA). In vitro, 2-PMPA robustly enhanced myelination in dorsal root ganglion (DRG) explants. In vivo, using a sciatic nerve crush injury model in aged mice, 2-PMPA accelerated remyelination, as evidenced by increased myelin sheath thickness and higher numbers of remyelinated axons. These findings suggest that GCPII inhibition may be a promising therapeutic strategy to enhance remyelination and potentially improve functional recovery after PNI, which is especially relevant in elderly PNI patients where this process is compromised.
Collapse
Affiliation(s)
- Yu Su
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (Y.S.); (M.H.); (A.G.T.); (J.M.); (K.D.J.H.); (Y.Z.); (Y.W.)
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Meixiang Huang
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (Y.S.); (M.H.); (A.G.T.); (J.M.); (K.D.J.H.); (Y.Z.); (Y.W.)
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ajit G. Thomas
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (Y.S.); (M.H.); (A.G.T.); (J.M.); (K.D.J.H.); (Y.Z.); (Y.W.)
| | - John Maragakis
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (Y.S.); (M.H.); (A.G.T.); (J.M.); (K.D.J.H.); (Y.Z.); (Y.W.)
| | - Kaitlyn D. J. Huizar
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (Y.S.); (M.H.); (A.G.T.); (J.M.); (K.D.J.H.); (Y.Z.); (Y.W.)
| | - Yuxin Zheng
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (Y.S.); (M.H.); (A.G.T.); (J.M.); (K.D.J.H.); (Y.Z.); (Y.W.)
| | - Ying Wu
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (Y.S.); (M.H.); (A.G.T.); (J.M.); (K.D.J.H.); (Y.Z.); (Y.W.)
| | - Mohamed H. Farah
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Barbara S. Slusher
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (Y.S.); (M.H.); (A.G.T.); (J.M.); (K.D.J.H.); (Y.Z.); (Y.W.)
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Psychiatry and Behavioral Science, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
7
|
Wiseman RL, Bigos KL, Dastgheyb RM, Barker PB, Rubin LH, Slusher BS. Brain N -acetyl-aspartyl-glutamate is associated with cognitive function in older virally suppressed people with HIV. AIDS 2024; 38:1003-1011. [PMID: 38411600 PMCID: PMC11062820 DOI: 10.1097/qad.0000000000003871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 02/05/2024] [Accepted: 02/14/2024] [Indexed: 02/28/2024]
Abstract
OBJECTIVES Cognitive impairment persists in virally suppressed people with HIV (VS-PWH) especially in higher order domains. One cortical circuit, linked to these domains, is regulated by N -acetyl-aspartyl glutamate (NAAG), the endogenous agonist of the metabotropic glutamate receptor 3. The enzyme glutamate carboxypeptidase II (GCPII) catabolizes NAAG and is upregulated in aging and disease. Inhibition of GCPII increases brain NAAG and improves learning and memory in rodent and primate models. DESIGN As higher order cognitive impairment is present in VS-PWH, and NAAG has not been investigated in earlier magnetic resonance spectroscopy studies (MRS), we investigated if brain NAAG levels measured by MRS were associated with cognitive function. METHODS We conducted a retrospective analysis of 7-Tesla MRS data from a previously published study on cognition in older VS-PWH. The original study did not separately quantify NAAG, therefore, work for this report focused on relationships between regional NAAG levels in frontal white matter (FWM), left hippocampus, left basal ganglia and domain-specific cognitive performance in 40 VS-PWH after adjusting for confounds. Participants were older than 50 years, negative for affective and neurologic disorders, and had no prior 3-month psychoactive-substance use. RESULTS Higher NAAG levels in FWM were associated with better attention/working memory. Higher left basal ganglia NAAG related to better verbal fluency. There was a positive relationship between hippocampal NAAG and executive function which lost significance after correction for confounds. CONCLUSION These data suggest brain NAAG serves as a biomarker of cognition in VS-PWH. Pharmacological modulation of brain NAAG warrants investigation as a therapeutic approach for cognitive deficits in VS-PWH.
Collapse
Affiliation(s)
- Robyn L. Wiseman
- Department of Pharmacology and Molecular Sciences
- Johns Hopkins Drug Discovery
- Department of Medicine, Division of Clinical Pharmacology
| | - Kristin L. Bigos
- Department of Pharmacology and Molecular Sciences
- Department of Medicine, Division of Clinical Pharmacology
- Department of Psychiatry and Behavioral Sciences
| | | | - Peter B. Barker
- Russell H. Morgan Department of Radiology and Radiological Sciences
| | - Leah H. Rubin
- Department of Psychiatry and Behavioral Sciences
- Department of Neurology
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health
| | - Barbara S. Slusher
- Department of Pharmacology and Molecular Sciences
- Johns Hopkins Drug Discovery
- Department of Medicine, Division of Clinical Pharmacology
- Department of Psychiatry and Behavioral Sciences
- Department of Neurology
- Department of Oncology
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
8
|
Arnsten AFT, Wang M, D’Esposito M. Dynamic Network Connectivity: from monkeys to humans. Front Hum Neurosci 2024; 18:1353043. [PMID: 38384333 PMCID: PMC10879414 DOI: 10.3389/fnhum.2024.1353043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 01/19/2024] [Indexed: 02/23/2024] Open
Abstract
Human brain imaging research using functional MRI (fMRI) has uncovered flexible variations in the functional connectivity between brain regions. While some of this variability likely arises from the pattern of information flow through circuits, it may also be influenced by rapid changes in effective synaptic strength at the molecular level, a phenomenon called Dynamic Network Connectivity (DNC) discovered in non-human primate circuits. These neuromodulatory molecular mechanisms are found in layer III of the macaque dorsolateral prefrontal cortex (dlPFC), the site of the microcircuits shown by Goldman-Rakic to be critical for working memory. This research has shown that the neuromodulators acetylcholine, norepinephrine, and dopamine can rapidly change the strength of synaptic connections in layer III dlPFC by (1) modifying the depolarization state of the post-synaptic density needed for NMDA receptor neurotransmission and (2) altering the open state of nearby potassium channels to rapidly weaken or strengthen synaptic efficacy and the strength of persistent neuronal firing. Many of these actions involve increased cAMP-calcium signaling in dendritic spines, where varying levels can coordinate the arousal state with the cognitive state. The current review examines the hypothesis that some of the dynamic changes in correlative strength between cortical regions observed in human fMRI studies may arise from these molecular underpinnings, as has been seen when pharmacological agents or genetic alterations alter the functional connectivity of the dlPFC consistent with the macaque physiology. These DNC mechanisms provide essential flexibility but may also confer vulnerability to malfunction when dysregulated in cognitive disorders.
Collapse
Affiliation(s)
- Amy F. T. Arnsten
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, United States
| | - Min Wang
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, United States
| | - Mark D’Esposito
- Department of Psychology, Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, United States
| |
Collapse
|
9
|
Ji T, Pang Y, Cheng M, Wang R, Chen X, Zhang C, Liu M, Zhang J, Zhong C. Deletion of glutamate carboxypeptidase II (GCPII), but not GCPIII, provided long-term benefits in mice with traumatic brain injury. CNS Neurosci Ther 2023; 29:3786-3801. [PMID: 37349952 PMCID: PMC10651966 DOI: 10.1111/cns.14299] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/19/2023] [Accepted: 05/28/2023] [Indexed: 06/24/2023] Open
Abstract
MAIN PROBLEM N-acetylaspartylglutamate (NAAG) has neuroprotective effects in traumatic brain injury (TBI) by activating metabotropic glutamate receptor 3 (mGluR3) and reducing glutamate release. Glutamate carboxypeptidase II (GCPII) is the primary enzyme responsible for the hydrolysis of NAAG. It remains unclear whether glutamate carboxypeptidase III (GCPIII), a homolog of GCPII, can partially compensate for GCPII's function. METHODS GCPII-/- , GCPIII-/- , and GCPII/III-/- mice were generated using CRISPR/Cas9 technology. Mice brain injury model was established through moderate controlled cortical impact (CCI). The relationship between GCPII and GCPIII was explored by analyzing injury response signals in the hippocampus and cortex of mice with different genotypes at the acute (1 day) and subacute (7 day) phase after TBI. RESULTS In this study, we found that deletion of GCPII reduced glutamate production, excitotoxicity, and neuronal damage and improved cognitive function, but GCPIII deletion had no significant neuroprotective effect. Additionally, there was no significant difference in the neuroprotective effect between the combination of GCPII and GCPIII deletion and GCPII deletion alone. CONCLUSION These results suggest that GCPII inhibition may be a therapeutic option for TBI, and that GCPIII may not act as a complementary enzyme to GCPII in this context.
Collapse
Affiliation(s)
- Tongjie Ji
- Department of NeurosurgeryShanghai East Hospital, School of Medicine, Tongji UniversityShanghaiChina
| | - Ying Pang
- Department of NeurosurgeryShanghai East Hospital, School of Medicine, Tongji UniversityShanghaiChina
| | - Meng Cheng
- Department of NeurosurgeryShanghai East Hospital, School of Medicine, Tongji UniversityShanghaiChina
| | - Rui Wang
- Department of NeurosurgeryShanghai East Hospital, School of Medicine, Tongji UniversityShanghaiChina
| | - Xu Chen
- Department of NeurosurgeryShanghai East Hospital, School of Medicine, Tongji UniversityShanghaiChina
| | - Chunyu Zhang
- Department of NeurosurgeryShanghai East Hospital, School of Medicine, Tongji UniversityShanghaiChina
| | - Min Liu
- Department of NeurosurgeryShanghai East Hospital, School of Medicine, Tongji UniversityShanghaiChina
| | - Jing Zhang
- Department of NeurosurgeryShanghai East Hospital, School of Medicine, Tongji UniversityShanghaiChina
- Institute for Advanced StudyTongji UniversityShanghaiChina
| | - Chunlong Zhong
- Department of NeurosurgeryShanghai East Hospital, School of Medicine, Tongji UniversityShanghaiChina
| |
Collapse
|
10
|
Arnsten AFT, Ishizawa Y, Xie Z. Scientific rationale for the use of α2A-adrenoceptor agonists in treating neuroinflammatory cognitive disorders. Mol Psychiatry 2023; 28:4540-4552. [PMID: 37029295 PMCID: PMC10080530 DOI: 10.1038/s41380-023-02057-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 03/22/2023] [Accepted: 03/24/2023] [Indexed: 04/09/2023]
Abstract
Neuroinflammatory disorders preferentially impair the higher cognitive and executive functions of the prefrontal cortex (PFC). This includes such challenging disorders as delirium, perioperative neurocognitive disorder, and the sustained cognitive deficits from "long-COVID" or traumatic brain injury. There are no FDA-approved treatments for these symptoms; thus, understanding their etiology is important for generating therapeutic strategies. The current review describes the molecular rationale for why PFC circuits are especially vulnerable to inflammation, and how α2A-adrenoceptor (α2A-AR) actions throughout the nervous and immune systems can benefit the circuits in PFC needed for higher cognition. The layer III circuits in the dorsolateral PFC (dlPFC) that generate and sustain the mental representations needed for higher cognition have unusual neurotransmission and neuromodulation. They are wholly dependent on NMDAR neurotransmission, with little AMPAR contribution, and thus are especially vulnerable to kynurenic acid inflammatory signaling which blocks NMDAR. Layer III dlPFC spines also have unusual neuromodulation, with cAMP magnification of calcium signaling in spines, which opens nearby potassium channels to rapidly weaken connectivity and reduce neuronal firing. This process must be tightly regulated, e.g. by mGluR3 or α2A-AR on spines, to prevent loss of firing. However, the production of GCPII inflammatory signaling reduces mGluR3 actions and markedly diminishes dlPFC network firing. Both basic and clinical studies show that α2A-AR agonists such as guanfacine can restore dlPFC network firing and cognitive function, through direct actions in the dlPFC, but also by reducing the activity of stress-related circuits, e.g. in the locus coeruleus and amygdala, and by having anti-inflammatory actions in the immune system. This information is particularly timely, as guanfacine is currently the focus of large clinical trials for the treatment of delirium, and in open label studies for the treatment of cognitive deficits from long-COVID.
Collapse
Affiliation(s)
- Amy F T Arnsten
- Department Neuroscience, Yale University School of Medicine, New Haven, CT, 056510, USA.
| | - Yumiko Ishizawa
- Department Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Zhongcong Xie
- Department Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| |
Collapse
|
11
|
Bathla S, Datta D, Liang F, Barthelemy N, Wiseman R, Slusher BS, Asher J, Zeiss C, Ekanayake‐Alper D, Holden D, Terwilliger G, Duque A, Arellano J, van Dyck C, Bateman RJ, Xie Z, Nairn AC, Arnsten AFT. Chronic GCPII (glutamate-carboxypeptidase-II) inhibition reduces pT217Tau levels in the entorhinal and dorsolateral prefrontal cortices of aged macaques. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2023; 9:e12431. [PMID: 37915375 PMCID: PMC10617575 DOI: 10.1002/trc2.12431] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/28/2023] [Accepted: 09/30/2023] [Indexed: 11/03/2023]
Abstract
Introduction Current approaches for treating sporadic Alzheimer's disease (sAD) focus on removal of amyloid beta 1-42 (Aβ1-42) or phosphorylated tau, but additional strategies are needed to reduce neuropathology at earlier stages prior to neuronal damage. Longstanding data show that calcium dysregulation is a key etiological factor in sAD, and the cortical neurons most vulnerable to tau pathology show magnified calcium signaling, for example in dorsolateral prefrontal cortex (dlPFC) and entorhinal cortex (ERC). In primate dlPFC and ERC, type 3 metabotropic glutamate receptors (mGluR3s) are predominately post-synaptic, on spines, where they regulate cAMP-calcium signaling, a process eroded by inflammatory glutamate carboxypeptidase II (GCPII) actions. The current study tested whether enhancing mGluR3 regulation of calcium via chronic inhibition of GCPII would reduce tau hyperphosphorylation in aged macaques with naturally-occurring tau pathology. Methods Aged rhesus macaques were treated daily with the GCPII inhibitor, 2-MPPA (2-3-mercaptopropyl-penanedioic acid (2-MPPA)),Aged rhesus macaques were treated daily with the GCPII inhibitor, 2-MPPA (2-3-mercaptopropyl-penanedioic acid (2-MPPA)). Results Aged macaques that received 2-MPPA had significantly lower pT217Tau levels in dlPFC and ERC, and had lowered plasma pT217Tau levels from baseline. pT217Tau levels correlated significantly with GCPII activity in dlPFC. Both 2-MPPA- and vehicle-treated monkeys showed cognitive improvement; 2-MPPA had no apparent side effects. Exploratory CSF analyses indicated reduced pS202Tau with 2-MPPA administration, confirmed in dlPFC samples. Discussion These data provide proof-of-concept support that GCPII inhibition can reduce tau hyperphosphorylation in the primate cortices most vulnerable in sAD. GCPII inhibition may be particularly helpful in reducing the risk of sAD caused by inflammation. These data in nonhuman primates should encourage future research on this promising mechanism. Highlights Inflammation is a key driver of sporadic Alzheimer's disease.GCPII inflammatory signaling in brain decreases mGluR3 regulation of calcium.Chronic inhibition of GCPII inflammatory signaling reduced pT217Tau in aged monkeys.GCPII inhibition is a novel strategy to help prevent tau pathology at early stages.
Collapse
Affiliation(s)
- Shveta Bathla
- Departments of PsychiatryYale University School of MedicineNew HavenConnecticutUSA
| | - Dibyadeep Datta
- Departments of PsychiatryYale University School of MedicineNew HavenConnecticutUSA
- Departments of NeuroscienceYale University School of MedicineNew HavenConnecticutUSA
| | - Feng Liang
- Department of AnesthesiologyHarvard University School of MedicineBostonMassachusettsUSA
| | - Nicolas Barthelemy
- Department of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
| | - Robyn Wiseman
- Department of Neurology, Johns Hopkins University Drug DiscoveryJohns Hopkins School of MedicineBaltimoreMarylandUSA
| | - Barbara S Slusher
- Department of Neurology, Johns Hopkins University Drug DiscoveryJohns Hopkins School of MedicineBaltimoreMarylandUSA
| | - Jennifer Asher
- Departments of Comparative MedicineYale University School of MedicineNew HavenConnecticutUSA
| | - Caroline Zeiss
- Departments of Comparative MedicineYale University School of MedicineNew HavenConnecticutUSA
| | - Dil Ekanayake‐Alper
- Departments of Comparative MedicineYale University School of MedicineNew HavenConnecticutUSA
| | - Daniel Holden
- Departments of RadiologyYale University School of MedicineNew HavenConnecticutUSA
| | - Gordon Terwilliger
- Departments of Comparative MedicineYale University School of MedicineNew HavenConnecticutUSA
| | - Alvaro Duque
- Departments of NeuroscienceYale University School of MedicineNew HavenConnecticutUSA
| | - Jon Arellano
- Departments of NeuroscienceYale University School of MedicineNew HavenConnecticutUSA
| | - Christopher van Dyck
- Departments of PsychiatryYale University School of MedicineNew HavenConnecticutUSA
| | - Randall J. Bateman
- Departments of RadiologyYale University School of MedicineNew HavenConnecticutUSA
| | - Zhongcong Xie
- Departments of Comparative MedicineYale University School of MedicineNew HavenConnecticutUSA
| | - Angus C. Nairn
- Departments of PsychiatryYale University School of MedicineNew HavenConnecticutUSA
| | - Amy F. T. Arnsten
- Departments of NeuroscienceYale University School of MedicineNew HavenConnecticutUSA
| |
Collapse
|
12
|
Faulkner MB, Rizk M, Bazzi Z, Dysko RC, Zhang Z. Sex-Specific Effects of Buprenorphine on Endoplasmic Reticulum Stress, Abnormal Protein Accumulation, and Cell Loss After Pediatric Mild Traumatic Brain Injury in Mice. Neurotrauma Rep 2023; 4:573-585. [PMID: 37752926 PMCID: PMC10518695 DOI: 10.1089/neur.2023.0051] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2023] Open
Abstract
Traumatic brain injury (TBI) in children often leads to poor developmental outcomes attributable to progressive cell loss caused by secondary injuries, including endoplasmic reticulum (ER) stress. Buprenorphine (BPN) is commonly used in children for pain management; however, the effects of BPN on ER stress in the pediatric population are still inconclusive. This study investigated the sex-specific effects of BPN on ER stress, abnormal protein accumulation, and cell loss in a mouse impact acceleration model of pediatric TBI. On post-natal day 20-21 (P20-21), male and female littermates were randomized into sham, TBI + saline and TBI + BPN groups. BPN (0.075 mg/kg) was administered to TBI + BPN mice at 30 min after injury and then every 6-12 h for 2 days. The impact of BPN was evaluated at 1, 3, and 7 days post-injury. We found that TBI induced more prominent ER stress pathway activation at 1 and 3 days post-injury in males, compared to females, whereas abnormal protein accumulation and cell loss were more severe in females at 7 days post-injury, compared with males. Although BPN partially ameliorated abnormal protein accumulation and cell loss in both males and females, BPN only decreased ER stress pathway activation in males, not in females. In conclusion, BPN exhibits sex-specific effects on ER stress, abnormal protein accumulation, and cell loss in a time-dependent manner at the acute phase after pediatric TBI, which provides the rationale to assess the potential effects of BPN on long-term outcomes after pediatric TBI in both males and females.
Collapse
Affiliation(s)
- Megan B. Faulkner
- Department of Natural Sciences, University of Michigan–Dearborn, Dearborn, Michigan, USA
| | - Mariam Rizk
- Department of Natural Sciences, University of Michigan–Dearborn, Dearborn, Michigan, USA
| | - Zahraa Bazzi
- Department of Natural Sciences, University of Michigan–Dearborn, Dearborn, Michigan, USA
| | - Robert C. Dysko
- Unit for Laboratory Animal Medicine, University of Michigan–Ann Arbor, Ann Arbor, Michigan, USA
| | - Zhi Zhang
- Department of Natural Sciences, University of Michigan–Dearborn, Dearborn, Michigan, USA
| |
Collapse
|
13
|
Datta D. Interrogating the Etiology of Sporadic Alzheimer's Disease Using Aging Rhesus Macaques: Cellular, Molecular, and Cortical Circuitry Perspectives. J Gerontol A Biol Sci Med Sci 2023; 78:1523-1534. [PMID: 37279946 PMCID: PMC10460555 DOI: 10.1093/gerona/glad134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Indexed: 06/08/2023] Open
Abstract
Aging is the most significant risk factor for neurodegenerative disorders such as Alzheimer's disease (AD) associated with profound socioeconomic and personal costs. Consequently, there is an urgent need for animal models that recapitulate the age-related spatial and temporal complexity and patterns of pathology identical to human AD. Our research in aging nonhuman primate models involving rhesus macaques has revealed naturally occurring amyloid and tau pathology, including the formation of amyloid plaques and neurofibrillary tangles comprising hyperphosphorylated tau. Moreover, rhesus macaques exhibit synaptic dysfunction in association cortices and cognitive impairments with advancing age, and thus can be used to interrogate the etiological mechanisms that generate neuropathological cascades in sporadic AD. Particularly, unique molecular mechanisms (eg, feedforward cyclic adenosine 3',5'-monophosphate [cAMP]-Protein kinase A (PKA)-calcium signaling) in the newly evolved primate dorsolateral prefrontal cortex are critical for persistent firing required for subserving higher-order cognition. For example, dendritic spines in primate dorsolateral prefrontal cortex contain a specialized repertoire of proteins to magnify feedforward cAMP-PKA-calcium signaling such as N-methyl-d-aspartic acid receptors and calcium channels on the smooth endoplasmic reticulum (eg, ryanodine receptors). This process is constrained by phosphodiesterases (eg, PDE4) that hydrolyze cAMP and calcium-buffering proteins (eg, calbindin) in the cytosol. However, genetic predispositions and age-related insults exacerbate feedforward cAMP-Protein kinase A-calcium signaling pathways that induce a myriad of downstream effects, including the opening of K+ channels to weaken network connectivity, calcium-mediated dysregulation of mitochondria, and activation of inflammatory cascades to eliminate synapses, thereby increasing susceptibility to atrophy. Therefore, aging rhesus macaques provide an invaluable model to explore novel therapeutic strategies in sporadic AD.
Collapse
Affiliation(s)
- Dibyadeep Datta
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
14
|
Zhang F, Zhang Z, Alt J, Kambhampati SP, Sharma A, Singh S, Nance E, Thomas AG, Rojas C, Rais R, Slusher BS, Kannan RM, Kannan S. Dendrimer-enabled targeted delivery attenuates glutamate excitotoxicity and improves motor function in a rabbit model of cerebral palsy. J Control Release 2023; 358:27-42. [PMID: 37054778 PMCID: PMC10330216 DOI: 10.1016/j.jconrel.2023.04.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 03/17/2023] [Accepted: 04/10/2023] [Indexed: 04/15/2023]
Abstract
Glutamate carboxypeptidase II (GCPII), localized on the surface of astrocytes and activated microglia, regulates extracellular glutamate concentration in the central nervous system (CNS). We have previously shown that GCPII is upregulated in activated microglia in the presence of inflammation. Inhibition of GCPII activity could reduce glutamate excitotoxicity, which may decrease inflammation and promote a 'normal' microglial phenotype. 2-(3-Mercaptopropyl) pentanedioic acid (2-MPPA) is the first GCPII inhibitor that underwent clinical trials. Unfortunately, immunological toxicities have hindered 2-MPPA clinical translation. Targeted delivery of 2-MPPA specifically to activated microglia and astrocytes that over-express GCPII has the potential to mitigate glutamate excitotoxicity and attenuate neuroinflammation. In this study, we demonstrate that 2-MPPA when conjugated to generation-4, hydroxyl-terminated polyamidoamine (PAMAM) dendrimers (D-2MPPA) localize specifically in activated microglia and astrocytes only in newborn rabbits with cerebral palsy (CP), not in controls. D-2MPPA treatment led to higher 2-MPPA levels in the injured brain regions compared to 2-MPPA treatment, and the extent of D-2MPPA uptake correlated with the injury severity. D-2MPPA was more efficacious than 2-MPPA in decreasing extracellular glutamate level in ex vivo brain slices of CP kits, and in increasing transforming growth factor beta 1 (TGF-β1) level in primary mixed glial cell cultures. A single systemic intravenous dose of D-2MPPA on postnatal day 1 (PND1) decreased microglial activation and resulted in a change in microglial morphology to a more ramified form along with amelioration of motor deficits by PND5. These results indicate that targeted dendrimer-based delivery specifically to activated microglia and astrocytes can improve the efficacy of 2-MPPA by attenuating glutamate excitotoxicity and microglial activation.
Collapse
Affiliation(s)
- Fan Zhang
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Zhi Zhang
- Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jesse Alt
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Siva P Kambhampati
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Anjali Sharma
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Sarabdeep Singh
- Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Elizabeth Nance
- Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ajit G Thomas
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Camilo Rojas
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Rana Rais
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Barbara S Slusher
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Rangaramanujam M Kannan
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| | - Sujatha Kannan
- Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
15
|
Sah N, Zhang Z, Chime A, Fowler A, Mendez-Trendler A, Sharma A, Kannan RM, Slusher B, Kannan S. Dendrimer-Conjugated Glutamate Carboxypeptidase II Inhibitor Restores Microglial Changes in a Rabbit Model of Cerebral Palsy. Dev Neurosci 2023; 45:268-275. [PMID: 36990069 PMCID: PMC10614263 DOI: 10.1159/000530389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 03/17/2023] [Indexed: 03/31/2023] Open
Abstract
We have previously shown that maternal endotoxin exposure leads to a phenotype of cerebral palsy and pro-inflammatory microglia in the brain in neonatal rabbits. "Activated" microglia overexpress the enzyme glutamate carboxypeptidase II (GCPII) that hydrolyzes N-acetylaspartylglutamate to N-acetylaspartate and glutamate, and we have shown previously that inhibiting microglial GCPII is neuroprotective. Glutamate-induced injury and associated immune signaling can alter microglial responses including microglial process movements for surveillance and phagocytosis. We hypothesize that inhibition of GCPII activity could alter microglial phenotype and normalize microglial process movement/dynamics. Newborn rabbit kits exposed to endotoxin in utero, when treated with dendrimer-conjugated 2-(phosphonomethyl)-pentanedioic acid (D-2PMPA), a potent and selective inhibitor of microglial GCPII, showed profound changes in microglial phenotype within 48 h of treatment. Live imaging of hippocampal microglia in ex vivo brain slice preparations revealed larger cell body and phagocytic cup sizes with less stable microglia processes in CP kits compared to healthy controls. D-2PMPA treatment led to significant reversal of microglial process stability to healthy control levels. Our results emphasize the importance of microglial process dynamics in determining the state of microglial function in the developing brain and demonstrate how GCPII inhibition specifically in microglia can effectively change the microglial process motility to healthy control levels, potentially impacting migration, phagocytosis, and inflammatory functions.
Collapse
Affiliation(s)
- Nirnath Sah
- Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zhi Zhang
- Department of Natural Sciences, University of Michigan-Dearborn, Dearborn, MI, USA
| | - Alicia Chime
- Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Amanda Fowler
- Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Antonio Mendez-Trendler
- Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anjali Sharma
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Chemistry, Washington State University, Pullman, WA, USA
| | - Rangaramanujam M. Kannan
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Barbara Slusher
- Johns Hopkins Drug Discovery Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sujatha Kannan
- Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
16
|
Glucagon-like Peptide 1 Receptor Activation Inhibits Microglial Pyroptosis via Promoting Mitophagy to Alleviate Depression-like Behaviors in Diabetic Mice. Nutrients 2022; 15:nu15010038. [PMID: 36615696 PMCID: PMC9824834 DOI: 10.3390/nu15010038] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/05/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Depression is a frequent and serious comorbidity associated with diabetes which adversely affects prognosis and quality of life. Glucagon-like peptide-1 receptor (GLP-1R) agonists, widely used in the treatment of diabetes, are reported to exert neuroprotective effects in the central nervous system. Thus, we aim to evaluate whether GLP-1R agonist exendin-4 (EX-4) could alleviate depression-like behaviors in diabetic mice and to explore its underlying mechanism. The antidepressant effects of EX-4 were evaluated using behavioral tests in db/db mice. The effects of EX-4 on microglial pyroptosis and neuroinflammation were assessed in N9 microglial cells. EX-4 administration alleviated depression-like behaviors in diabetic db/db mice. GLP-1R activation by EX-4 significantly suppressed microglial pyroptosis and neuroinflammation by downregulation of gasdermin D (GSDMD) and interleukin (IL)-1β in diabetic mice and lipopolysaccharide (LPS)-primed N9 microglia. Mechanistically, GLP-1R activation improved mitochondrial function and promoted mitophagy by decreasing the accumulation of mitochondrial reactive oxygen species (mtROS) and intracellular ROS production. EX-4 exhibits antidepressant effects in depression associated with diabetes in diabetic mice, which may be mediated by inhibiting microglial pyroptisis via promoting mitophagy. It is supposed that GLP-1R agonists may be a promising therapy in depression associated with diabetes.
Collapse
|
17
|
Arnsten AFT, Woo E, Yang S, Wang M, Datta D. Unusual Molecular Regulation of Dorsolateral Prefrontal Cortex Layer III Synapses Increases Vulnerability to Genetic and Environmental Insults in Schizophrenia. Biol Psychiatry 2022; 92:480-490. [PMID: 35305820 PMCID: PMC9372235 DOI: 10.1016/j.biopsych.2022.02.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 02/03/2022] [Accepted: 02/06/2022] [Indexed: 02/06/2023]
Abstract
Schizophrenia is associated with reduced numbers of spines and dendrites from layer III of the dorsolateral prefrontal cortex (dlPFC), the layer that houses the recurrent excitatory microcircuits that subserve working memory and abstract thought. Why are these synapses so vulnerable, while synapses in deeper or more superficial layers are little affected? This review describes the special molecular properties that govern layer III neurotransmission and neuromodulation in the primate dlPFC and how they may render these circuits particularly vulnerable to genetic and environmental insults. These properties include a reliance on NMDA receptor rather than AMPA receptor neurotransmission; cAMP (cyclic adenosine monophosphate) magnification of calcium signaling near the glutamatergic synapse of dendritic spines; and potassium channels opened by cAMP/PKA (protein kinase A) signaling that dynamically alter network strength, with built-in mechanisms to take dlPFC "offline" during stress. A variety of genetic and/or environmental insults can lead to the same phenotype of weakened layer III connectivity, in which mechanisms that normally strengthen connectivity are impaired and those that normally weaken connectivity are intensified. Inflammatory mechanisms, such as increased kynurenic acid and glutamate carboxypeptidase II expression, are especially detrimental to layer III dlPFC neurotransmission and modulation, mimicking genetic insults. The combination of genetic and inflammatory insults may cross the threshold into pathology.
Collapse
Affiliation(s)
- Amy F T Arnsten
- Department of Neuroscience, Yale Medical School, New Haven, Connecticut.
| | - Elizabeth Woo
- Department of Neuroscience, Yale Medical School, New Haven, Connecticut
| | - Shengtao Yang
- Department of Neuroscience, Yale Medical School, New Haven, Connecticut
| | - Min Wang
- Department of Neuroscience, Yale Medical School, New Haven, Connecticut
| | - Dibyadeep Datta
- Department of Neuroscience, Yale Medical School, New Haven, Connecticut
| |
Collapse
|
18
|
Gonzalez A, Hammock EAD. Oxytocin and microglia in the development of social behaviour. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210059. [PMID: 35858111 PMCID: PMC9272152 DOI: 10.1098/rstb.2021.0059] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 04/18/2022] [Indexed: 08/31/2023] Open
Abstract
Oxytocin is a well-established regulator of social behaviour. Microglia, the resident immune cells of the central nervous system, regulate brain development and maintenance in health and disease. Oxytocin and microglia interact: microglia appear to regulate the oxytocin system and are, in turn, regulated by oxytocin, which appears to have anti-inflammatory effects. Both microglia and oxytocin are regulated in sex-specific ways. Oxytocin and microglia may work together to promote experience-dependent circuit refinement through multiple developmental-sensitive periods contributing to individual differences in social behaviour. This article is part of the theme issue 'Interplays between oxytocin and other neuromodulators in shaping complex social behaviours'.
Collapse
Affiliation(s)
- Alicia Gonzalez
- Department of Psychology and Program in Neuroscience, Florida State University, 1107 West Call Street, Tallahassee, FL 32306, USA
| | - Elizabeth A. D. Hammock
- Department of Psychology and Program in Neuroscience, Florida State University, 1107 West Call Street, Tallahassee, FL 32306, USA
| |
Collapse
|
19
|
Characterization of a mGluR5 Knockout Rat Model with Hallmarks of Fragile X Syndrome. Life (Basel) 2022; 12:life12091308. [PMID: 36143345 PMCID: PMC9504063 DOI: 10.3390/life12091308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022] Open
Abstract
The number of reported cases of neurodevelopmental disorders has increased significantly in the last few decades, but the etiology of these diseases remains poorly understood. There is evidence of a fundamental link between genetic abnormalities and symptoms of autism spectrum disorders (ASDs), and the most common monogenetic inheritable form of ASDs is Fragile X Syndrome (FXS). Previous studies indicate that FXS is linked to glutamate signaling regulation by the G-protein-coupled metabotropic glutamate receptor 5 (mGluR5), which has been shown to have a regulatory role in neuroinflammation. We characterized the effect of knocking out mGluR5 in an organism known to have complex cognitive functions—the rat. The heterozygous phenotype is the most clinically relevant; therefore, we performed analysis in heterozygous pups. We showed developmental abnormalities in heterozygous mGluR5 knockout rats, as well as a significant increase in chemokine (C-X-C motif) ligand 1 (CXCL) expression, a hallmark indicator of early onset inflammation. We quantified an increase in microglial density in the knockout pups and quantified morphological phenotypes representative of greater reactivity in the male vs. female and postnatal day 28 heterozygous pups compared to postnatal day 14 heterozygous pups. In response to injury, reactive microglia release matrix metalloproteases, contribute to extracellular matrix (ECM) breakdown, and are responsible for eradicating cellular and molecular debris. In our study, the changes in microglial density and reactivity correlated with abnormalities in the mRNA expression levels of ECM proteins and with the density of perineuronal nets. We saw atypical neuropsychiatric behavior in open field and elevated plus tests in heterozygous pups compared to wild-type litter and age-matched controls. These results demonstrate the pathological potential of the mGluR5 knockout in rats and further support the presence of neuroinflammatory roots in ASDs.
Collapse
|
20
|
Sex specific effects of buprenorphine on behavior, astrocytic opioid receptor expression and neuroinflammation after pediatric traumatic brain injury in mice. Brain Behav Immun Health 2022; 22:100469. [PMID: 35620644 PMCID: PMC9127176 DOI: 10.1016/j.bbih.2022.100469] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/29/2022] [Accepted: 05/06/2022] [Indexed: 12/31/2022] Open
Abstract
Children who suffered traumatic brain injury (TBI) often experience acute and chronic pain, which is linked to a poor quality of life. Buprenorphine (BPN) is commonly used to treat moderate to severe persistent pain in children, however, the efficacy and safety profile of BPN in the pediatric population is still inconclusive. This study investigated the sex-specific effects of BPN on body weight, motor coordination and strength, expression of opioid receptors in the white matter astrocytes, and neuroinflammation in a mouse impact acceleration model of pediatric TBI. Male and female littermates were randomized on postnatal day 20-21(P20-21) into Sham, TBI + saline and TBI + BPN groups. Mice in the TBI + saline and TBI + BPN groups underwent TBI, while the Sham group underwent anesthesia without injury. BPN (0.075 mg/kg) was administered to the TBI + BPN mice at 30 min after injury, and then every 6-12 h for 2 days. Mice in the TBI + saline group received the same amount of saline injections. The impact of BPN on body weight, motor function, opioid receptor expression, and neuroinflammation was evaluated at 1-day (d), 3-d and 7-d post-injury. We found that 1) TBI induced significant weight loss in both males and females. BPN treatment improved weight loss at 3-d post-injury in females. 2) TBI significantly impaired motor coordination and strength. BPN improved motor coordination and strength in both males and females at 1-d and 3-d post-injury. 3) TBI significantly decreased exploration activity at 1-d post-injury in males, and at 7-d post-injury in females, while BPN improved the exploration activity in females. 4) TBI significantly increased mRNA expression of mu-opioid receptors (MOR) at 7-d post-injury in males, but decreased mRNA expression of MOR at 1-d post-injury in females. BPN normalized MOR mRNA expression at 1-d post-injury in females. 5) MOR expression in astrocytes at corpus callosum significantly increased at 7-d post-injury in male TBI group, but significantly decreased at 1-d post-injury in female TBI group. BPN normalized MOR expression in both males and females. 6) TBI significantly increased the mRNA expression of TNF-α, IL-1β, IL-6 and iNOS. BPN decreased mRNA expression of iNOS, and increased mRNA expression of TGF-β1. In conclusion, this study elucidates the sex specific effects of BPN during the acute phase after pediatric TBI, which provides the rationale to assess potential effects of BPN on chronic pathological progressions after pediatric TBI in both males and females.
Collapse
|
21
|
Jeitner TM, Babich JW, Kelly JM. Advances in PSMA theranostics. Transl Oncol 2022; 22:101450. [PMID: 35597190 PMCID: PMC9123266 DOI: 10.1016/j.tranon.2022.101450] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/04/2022] [Accepted: 05/08/2022] [Indexed: 12/15/2022] Open
Abstract
PSMA is an appealing target for theranostic because it is a transmembrane protein with a known substrate that is overexpessed on prostate cancer cells and internalizes upon ligand binding. There are a number of PSMA theranostic ligands in clinical evaluation, clinical trial, or clinically approved. PSMA theranostic ligands increase progression-free survival, overall survival, and pain in patients with metastatic castration resistant prostate cancer. A major obstacle to PSMA-targeted radioligand therapy is off-target toxicity in salivary glands.
The validation of prostate specific membrane antigen (PSMA) as a molecular target in metastatic castration-resistant prostate cancer has stimulated the development of multiple classes of theranostic ligands that specifically target PSMA. Theranostic ligands are used to image disease or selectively deliver cytotoxic radioactivity to cells expressing PSMA according to the radioisotope conjugated to the ligand. PSMA theranostics is a rapidly advancing field that is now integrating into clinical management of prostate cancer patients. In this review we summarize published research describing the biological role(s) and activity of PSMA, highlight the most clinically advanced PSMA targeting molecules and biomacromolecules, and identify next generation PSMA ligands that aim to further improve treatment efficacy. The goal of this review is to provide a comprehensive assessment of the current state-of-play and a roadmap to achieving further advances in PSMA theranostics.
Collapse
Affiliation(s)
- Thomas M Jeitner
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, Belfer Research Building, 413 East 69th Street, Room BB-1604, New York, NY 10021, USA
| | - John W Babich
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, Belfer Research Building, 413 East 69th Street, Room BB-1604, New York, NY 10021, USA; Weill Cornell Medicine, Sandra and Edward Meyer Cancer Center, New York, NY 10021, USA; Weill Cornell Medicine, Citigroup Biomedical Imaging Center, New York, NY 10021, USA
| | - James M Kelly
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, Belfer Research Building, 413 East 69th Street, Room BB-1604, New York, NY 10021, USA; Weill Cornell Medicine, Citigroup Biomedical Imaging Center, New York, NY 10021, USA.
| |
Collapse
|
22
|
Morland C, Nordengen K. N-Acetyl-Aspartyl-Glutamate in Brain Health and Disease. Int J Mol Sci 2022; 23:ijms23031268. [PMID: 35163193 PMCID: PMC8836185 DOI: 10.3390/ijms23031268] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/20/2022] [Accepted: 01/21/2022] [Indexed: 02/04/2023] Open
Abstract
N-acetyl-aspartyl-glutamate (NAAG) is the most abundant dipeptide in the brain, where it acts as a neuromodulator of glutamatergic synapses by activating presynaptic metabotropic glutamate receptor 3 (mGluR3). Recent data suggest that NAAG is selectively localized to postsynaptic dendrites in glutamatergic synapses and that it works as a retrograde neurotransmitter. NAAG is released in response to glutamate and provides the postsynaptic neuron with a feedback mechanisms to inhibit excessive glutamate signaling. A key regulator of synaptically available NAAG is rapid degradation by the extracellular enzyme glutamate carboxypeptidase II (GCPII). Increasing endogenous NAAG—for instance by inhibiting GCPII—is a promising treatment option for many brain disorders where glutamatergic excitotoxicity plays a role. The main effect of NAAG occurs through increased mGluR3 activation and thereby reduced glutamate release. In the present review, we summarize the transmitter role of NAAG and discuss the involvement of NAAG in normal brain physiology. We further present the suggested roles of NAAG in various neurological and psychiatric diseases and discuss the therapeutic potential of strategies aiming to enhance NAAG levels.
Collapse
Affiliation(s)
- Cecilie Morland
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, The Faculty of Mathematics and Natural Sciences, University of Oslo, 1068 Oslo, Norway
- Correspondence: (C.M.); (K.N.); Tel.: +47-22844937; (C.M.); +47-23073580 (K.N.)
| | - Kaja Nordengen
- Department of Neurology, Oslo University Hospital, 0424 Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0318 Oslo, Norway
- Correspondence: (C.M.); (K.N.); Tel.: +47-22844937; (C.M.); +47-23073580 (K.N.)
| |
Collapse
|
23
|
Hollinger KR, Sharma A, Tallon C, Lovell L, Thomas AG, Zhu X, Wiseman R, Wu Y, Kambhampati SP, Liaw K, Sharma R, Rojas C, Rais R, Kannan S, Kannan RM, Slusher BS. Dendrimer-2PMPA selectively blocks upregulated microglial GCPII activity and improves cognition in a mouse model of multiple sclerosis. Nanotheranostics 2022; 6:126-142. [PMID: 34976589 PMCID: PMC8671953 DOI: 10.7150/ntno.63158] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/04/2021] [Indexed: 12/19/2022] Open
Abstract
Cognitive impairment is a common aspect of multiple sclerosis (MS) for which there are no treatments. Reduced brain N-acetylaspartylglutamate (NAAG) levels are linked to impaired cognition in various neurological diseases, including MS. NAAG levels are regulated by glutamate carboxypeptidase II (GCPII), which hydrolyzes the neuropeptide to N-acetyl-aspartate and glutamate. GCPII activity is upregulated multifold in microglia following neuroinflammation. Although several GCPII inhibitors, such as 2-PMPA, elevate brain NAAG levels and restore cognitive function in preclinical studies when given at high systemic doses or via direct brain injection, none are clinically available due to poor bioavailability and limited brain penetration. Hydroxyl-dendrimers have been successfully used to selectively deliver drugs to activated glia. Methods: We attached 2-PMPA to hydroxyl polyamidoamine (PAMAM) dendrimers (D-2PMPA) using a click chemistry approach. Cy5-labelled-D-2PMPA was used to visualize selective glial uptake in vitro and in vivo. D-2PMPA was evaluated for anti-inflammatory effects in LPS-treated glial cultures. In experimental autoimmune encephalomyelitis (EAE)-immunized mice, D-2PMPA was dosed biweekly starting at disease onset and cognition was assessed using the Barnes maze, and GCPII activity was measured in CD11b+ hippocampal cells. Results: D-2PMPA showed preferential uptake into microglia and robust anti-inflammatory activity, including elevations in NAAG, TGFβ, and mGluR3 in glial cultures. D-2PMPA significantly improved cognition in EAE mice, even though physical severity was unaffected. GCPII activity increased >20-fold in CD11b+ cells from EAE mice, which was significantly mitigated by D-2PMPA treatment. Conclusions: Hydroxyl dendrimers facilitate targeted drug delivery to activated microglia. These data support further development of D-2PMPA to attenuate elevated microglial GCPII activity and treat cognitive impairment in MS.
Collapse
Affiliation(s)
| | - Anjali Sharma
- Center for Nanomedicine, Department of Ophthalmology, Johns Hopkins University, Baltimore, MD, USA
| | - Carolyn Tallon
- Johns Hopkins Drug Discovery, Johns Hopkins University, Baltimore, MD, USA.,Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Lyndah Lovell
- Johns Hopkins Drug Discovery, Johns Hopkins University, Baltimore, MD, USA
| | - Ajit G Thomas
- Johns Hopkins Drug Discovery, Johns Hopkins University, Baltimore, MD, USA
| | - Xiaolei Zhu
- Johns Hopkins Drug Discovery, Johns Hopkins University, Baltimore, MD, USA.,Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Robyn Wiseman
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Ying Wu
- Johns Hopkins Drug Discovery, Johns Hopkins University, Baltimore, MD, USA
| | - Siva P Kambhampati
- Center for Nanomedicine, Department of Ophthalmology, Johns Hopkins University, Baltimore, MD, USA
| | - Kevin Liaw
- Center for Nanomedicine, Department of Ophthalmology, Johns Hopkins University, Baltimore, MD, USA.,Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Rishi Sharma
- Center for Nanomedicine, Department of Ophthalmology, Johns Hopkins University, Baltimore, MD, USA
| | - Camilo Rojas
- Johns Hopkins Drug Discovery, Johns Hopkins University, Baltimore, MD, USA.,Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Rana Rais
- Johns Hopkins Drug Discovery, Johns Hopkins University, Baltimore, MD, USA.,Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Sujatha Kannan
- Center for Nanomedicine, Department of Ophthalmology, Johns Hopkins University, Baltimore, MD, USA.,Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA.,Kennedy Krieger Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Rangaramanujam M Kannan
- Center for Nanomedicine, Department of Ophthalmology, Johns Hopkins University, Baltimore, MD, USA.,Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.,Kennedy Krieger Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Barbara S Slusher
- Johns Hopkins Drug Discovery, Johns Hopkins University, Baltimore, MD, USA.,Department of Neurology, Johns Hopkins University, Baltimore, MD, USA.,Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA.,Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, MD, USA.,Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA.,Department of Medicine, Johns Hopkins University, Baltimore, MD, USA.,Department of Oncology, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
24
|
Cools R, Arnsten AFT. Neuromodulation of prefrontal cortex cognitive function in primates: the powerful roles of monoamines and acetylcholine. Neuropsychopharmacology 2022; 47:309-328. [PMID: 34312496 PMCID: PMC8617291 DOI: 10.1038/s41386-021-01100-8] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 07/06/2021] [Accepted: 07/06/2021] [Indexed: 02/07/2023]
Abstract
The primate prefrontal cortex (PFC) subserves our highest order cognitive operations, and yet is tremendously dependent on a precise neurochemical environment for proper functioning. Depletion of noradrenaline and dopamine, or of acetylcholine from the dorsolateral PFC (dlPFC), is as devastating as removing the cortex itself, and serotonergic influences are also critical to proper functioning of the orbital and medial PFC. Most neuromodulators have a narrow inverted U dose response, which coordinates arousal state with cognitive state, and contributes to cognitive deficits with fatigue or uncontrollable stress. Studies in monkeys have revealed the molecular signaling mechanisms that govern the generation and modulation of mental representations by the dlPFC, allowing dynamic regulation of network strength, a process that requires tight regulation to prevent toxic actions, e.g., as occurs with advanced age. Brain imaging studies in humans have observed drug and genotype influences on a range of cognitive tasks and on PFC circuit functional connectivity, e.g., showing that catecholamines stabilize representations in a baseline-dependent manner. Research in monkeys has already led to new treatments for cognitive disorders in humans, encouraging future research in this important field.
Collapse
Affiliation(s)
- Roshan Cools
- Department of Psychiatry, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Amy F T Arnsten
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
25
|
Tallon C, Sharma A, Zhang Z, Thomas AG, Ng J, Zhu X, Donoghue A, Schulte M, Joe TR, Kambhampati SP, Sharma R, Liaw K, Kannan S, Kannan RM, Slusher BS. Dendrimer-2PMPA Delays Muscle Function Loss and Denervation in a Murine Model of Amyotrophic Lateral Sclerosis. Neurotherapeutics 2022; 19:274-288. [PMID: 34984651 PMCID: PMC9130402 DOI: 10.1007/s13311-021-01159-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/05/2021] [Indexed: 01/03/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease where muscle weakness and neuromuscular junction (NMJ) denervation precede motor neuron cell death. Although acetylcholine is the canonical neurotransmitter at the mammalian NMJ synapse, glutamate has recently been identified as a critical neurotransmitter for NMJ development and maintenance. One source of glutamate is through the catabolism of N-acetyl-aspartyl-glutamate (NAAG), which is found in mM concentrations in mammalian motoneurons, where it is released upon stimulation and hydrolyzed to glutamate by the glial enzyme glutamate carboxypeptidase II (GCPII). Using the SOD1G93A model of ALS, we found an almost fourfold elevation of GCPII enzymatic activity in SOD1G93A versus WT muscle and a robust increase in GCPII expression which was specifically associated with activated macrophages infiltrating the muscle. 2-(Phosphonomethyl)pentanedioic acid (2PMPA) is a potent GCPII inhibitor which robustly blocks glutamate release from NAAG but is highly polar with limited tissue penetration. To improve this, we covalently attached 2PMPA to a hydroxyl polyamidoamine (PAMAM-G4-OH) dendrimer delivery system (D-2PMPA) which is known to target activated macrophages in affected tissues. Systemic D-2PMPA therapy (20 mg/kg 2PMPA equivalent; IP 2 × /week) was found to localize in muscle macrophages in SOD1G93A mice and completely normalize the enhanced GCPII activity. Although no changes in body weight or survival were observed, D-2PMPA significantly improved grip strength and inhibited the loss of NMJ innervation in the gastrocnemius muscles. Our finding that inhibiting elevated GCPII activity in SOD1G93A muscle can prolong muscle function and delay NMJ denervation may have early therapeutic implications for ALS patients.
Collapse
Affiliation(s)
- Carolyn Tallon
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Anjali Sharma
- Center for Nanomedicine-Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Zhi Zhang
- Center for Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, USA
- Department of Natural Sciences, University of Michigan-Dearborn, Dearborn, MI, 48128, USA
| | - Ajit G Thomas
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Justin Ng
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Xiaolei Zhu
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, USA
- Department of Psychiatry and Behavioral Science, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Amanda Donoghue
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Michael Schulte
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Tawnjerae R Joe
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, USA
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Siva P Kambhampati
- Center for Nanomedicine-Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Rishi Sharma
- Center for Nanomedicine-Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Kevin Liaw
- Center for Nanomedicine-Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Sujatha Kannan
- Center for Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, USA
- Hugo W. Moser Research Institute at Kennedy-Krieger, Inc, Baltimore, USA
| | - Rangaramanujam M Kannan
- Center for Nanomedicine-Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, USA
- Hugo W. Moser Research Institute at Kennedy-Krieger, Inc, Baltimore, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Barbara S Slusher
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, USA.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, USA.
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, USA.
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, USA.
- Department of Psychiatry and Behavioral Science, Johns Hopkins University School of Medicine, Baltimore, USA.
- Johns Hopkins University School of Medicine, 855 N. Wolfe Street, Rangos 278, Baltimore, MD, 21205, USA.
| |
Collapse
|
26
|
Yang S, Datta D, Elizabeth Woo, Duque A, Morozov YM, Arellano J, Slusher BS, Wang M, Arnsten AFT. Inhibition of glutamate-carboxypeptidase-II in dorsolateral prefrontal cortex: potential therapeutic target for neuroinflammatory cognitive disorders. Mol Psychiatry 2022; 27:4252-4263. [PMID: 35732693 PMCID: PMC9718677 DOI: 10.1038/s41380-022-01656-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 05/27/2022] [Indexed: 02/07/2023]
Abstract
Glutamate carboxypeptidase-II (GCPII) expression in brain is increased by inflammation, e.g. by COVID19 infection, where it reduces NAAG stimulation of metabotropic glutamate receptor type 3 (mGluR3). GCPII-mGluR3 signaling is increasingly linked to higher cognition, as genetic alterations that weaken mGluR3 or increase GCPII signaling are associated with impaired cognition in humans. Recent evidence from macaque dorsolateral prefrontal cortex (dlPFC) shows that mGluR3 are expressed on dendritic spines, where they regulate cAMP-PKA opening of potassium (K+) channels to enhance neuronal firing during working memory. However, little is known about GCPII expression and function in the primate dlPFC, despite its relevance to inflammatory disorders. The present study used multiple label immunofluorescence and immunoelectron microscopy to localize GCPII in aging macaque dlPFC, and examined the effects of GCPII inhibition on dlPFC neuronal physiology and working memory function. GCPII was observed in astrocytes as expected, but also on neurons, including extensive expression in dendritic spines. Recordings in dlPFC from aged monkeys performing a working memory task found that iontophoresis of the GCPII inhibitors 2-MPPA or 2-PMPA markedly increased working memory-related neuronal firing and spatial tuning, enhancing neural representations. These beneficial effects were reversed by an mGluR2/3 antagonist, or by a cAMP-PKA activator, consistent with mGluR3 inhibition of cAMP-PKA-K+ channel signaling. Systemic administration of the brain penetrant inhibitor, 2-MPPA, significantly improved working memory performance without apparent side effects, with largest effects in the oldest monkeys. Taken together, these data endorse GCPII inhibition as a potential strategy for treating cognitive disorders associated with aging and/or neuroinflammation.
Collapse
Affiliation(s)
- Shengtao Yang
- grid.47100.320000000419368710Department Neuroscience, Yale University School of Medicine, New Haven, CT USA
| | - Dibyadeep Datta
- grid.47100.320000000419368710Department Neuroscience, Yale University School of Medicine, New Haven, CT USA ,grid.47100.320000000419368710Department Psychiatry, Yale University School of Medicine, New Haven, CT USA
| | - Elizabeth Woo
- grid.47100.320000000419368710Department Neuroscience, Yale University School of Medicine, New Haven, CT USA
| | - Alvaro Duque
- grid.47100.320000000419368710Department Neuroscience, Yale University School of Medicine, New Haven, CT USA
| | - Yury M. Morozov
- grid.47100.320000000419368710Department Neuroscience, Yale University School of Medicine, New Haven, CT USA
| | - Jon Arellano
- grid.47100.320000000419368710Department Neuroscience, Yale University School of Medicine, New Haven, CT USA
| | - Barbara S. Slusher
- grid.21107.350000 0001 2171 9311Department Neurology and Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD USA
| | - Min Wang
- grid.47100.320000000419368710Department Neuroscience, Yale University School of Medicine, New Haven, CT USA
| | - Amy F. T. Arnsten
- grid.47100.320000000419368710Department Neuroscience, Yale University School of Medicine, New Haven, CT USA
| |
Collapse
|
27
|
Krečmerová M, Majer P, Rais R, Slusher BS. Phosphonates and Phosphonate Prodrugs in Medicinal Chemistry: Past Successes and Future Prospects. Front Chem 2022; 10:889737. [PMID: 35668826 PMCID: PMC9163707 DOI: 10.3389/fchem.2022.889737] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/26/2022] [Indexed: 12/25/2022] Open
Abstract
Compounds with a phosphonate group, i.e., -P(O)(OH)2 group attached directly to the molecule via a P-C bond serve as suitable non-hydrolyzable phosphate mimics in various biomedical applications. In principle, they often inhibit enzymes utilizing various phosphates as substrates. In this review we focus mainly on biologically active phosphonates that originated from our institute (Institute of Organic Chemistry and Biochemistry in Prague); i.e., acyclic nucleoside phosphonates (ANPs, e.g., adefovir, tenofovir, and cidofovir) and derivatives of non-nucleoside phosphonates such as 2-(phosphonomethyl) pentanedioic acid (2-PMPA). Principal strategies of their syntheses and modifications to prodrugs is reported. Besides clinically used ANP antivirals, a special attention is paid to new biologically active molecules with respect to emerging infections and arising resistance of many pathogens against standard treatments. These new structures include 2,4-diamino-6-[2-(phosphonomethoxy)ethoxy]pyrimidines or so-called "open-ring" derivatives, acyclic nucleoside phosphonates with 5-azacytosine as a base moiety, side-chain fluorinated ANPs, aza/deazapurine ANPs. When transformed into an appropriate prodrug by derivatizing their charged functionalities, all these compounds show promising potential to become drug candidates for the treatment of viral infections. ANP prodrugs with suitable pharmacokinetics include amino acid phosphoramidates, pivaloyloxymethyl (POM) and isopropoxycarbonyloxymethyl (POC) esters, alkyl and alkoxyalkyl esters, salicylic esters, (methyl-2-oxo-1,3-dioxol-4-yl) methyl (ODOL) esters and peptidomimetic prodrugs. We also focus on the story of cytostatics related to 9-[2-(phosphonomethoxy)ethyl]guanine and its prodrugs which eventually led to development of the veterinary drug rabacfosadine. Various new ANP structures are also currently investigated as antiparasitics, especially antimalarial agents e.g., guanine and hypoxanthine derivatives with 2-(phosphonoethoxy)ethyl moiety, their thia-analogues and N-branched derivatives. In addition to ANPs and their analogs, we also describe prodrugs of 2-(phosphonomethyl)pentanedioic acid (2-PMPA), a potent inhibitor of the enzyme glutamate carboxypeptidase II (GCPII), also known as prostate-specific membrane antigen (PSMA). Glutamate carboxypeptidase II inhibitors, including 2-PMPA have been found efficacious in various preclinical models of neurological disorders which are caused by glutamatergic excitotoxicity. Unfortunately its highly polar character and hence low bioavailability severely limits its potential for clinical use. To overcome this problem, various prodrug strategies have been used to mask carboxylates and/or phosphonate functionalities with pivaloyloxymethyl, POC, ODOL and alkyl esters. Chemistry and biological characterization led to identification of prodrugs with 44-80 fold greater oral bioavailability (tetra-ODOL-2-PMPA).
Collapse
Affiliation(s)
- Marcela Krečmerová
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic (ASCR), Prague, Czechia
- *Correspondence: Marcela Krečmerová,
| | - Pavel Majer
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic (ASCR), Prague, Czechia
| | - Rana Rais
- Departments of Neurology, Pharmacology and Molecular Sciences, Johns Hopkins Drug Discovery, Baltimore, MD, United States
| | - Barbara S. Slusher
- Departments of Neurology, Pharmacology and Molecular Sciences, Psychiatry and Behavioral Sciences, Neuroscience, Medicine, Oncology, Johns Hopkins Drug Discovery, Baltimore, MD, United States
| |
Collapse
|
28
|
Datta D, Leslie SN, Woo E, Amancharla N, Elmansy A, Lepe M, Mecca AP, Slusher BS, Nairn AC, Arnsten AFT. Glutamate Carboxypeptidase II in Aging Rat Prefrontal Cortex Impairs Working Memory Performance. Front Aging Neurosci 2021; 13:760270. [PMID: 34867287 PMCID: PMC8634091 DOI: 10.3389/fnagi.2021.760270] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 09/28/2021] [Indexed: 11/29/2022] Open
Abstract
Glutamate carboxypeptidase II (GCPII) expression in brain is increased by inflammation, and reduces NAAG (N-acetyl aspartyl glutamate) stimulation of mGluR3 signaling. Genetic insults in this signaling cascade are increasingly linked to cognitive disorders in humans, where increased GCPII and or decreased NAAG-mGluR3 are associated with impaired prefrontal cortical (PFC) activation and cognitive impairment. As aging is associated with increased inflammation and PFC cognitive deficits, the current study examined GCPII and mGluR3 expression in the aging rat medial PFC, and tested whether GCPII inhibition with 2-(3-mercaptopropyl) pentanedioic acid (2-MPPA) would improve working memory performance. We found that GCPII protein was expressed on astrocytes and some microglia as expected from previous studies, but was also prominently expressed on neurons, and showed increased levels with advancing age. Systemic administration of the GCPII inhibitor, 2-MPPA, improved working memory performance in young and aged rats, and also improved performance after local infusion into the medial PFC. As GCPII inhibitors are well-tolerated, they may provide an important new direction for treatment of cognitive disorders associated with aging and/or inflammation.
Collapse
Affiliation(s)
- Dibyadeep Datta
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, United States
| | - Shannon N Leslie
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Elizabeth Woo
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, United States
| | - Nishita Amancharla
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, United States
| | - Ayah Elmansy
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, United States
| | - Miguel Lepe
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, United States
| | - Adam P Mecca
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Barbara S Slusher
- Department of Neurology and Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Angus C Nairn
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Amy F T Arnsten
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
29
|
Arnsten AFT, Datta D, Preuss TM. Studies of aging nonhuman primates illuminate the etiology of early-stage Alzheimer's-like neuropathology: An evolutionary perspective. Am J Primatol 2021; 83:e23254. [PMID: 33960505 PMCID: PMC8550995 DOI: 10.1002/ajp.23254] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 03/02/2021] [Accepted: 03/04/2021] [Indexed: 11/17/2022]
Abstract
Tau pathology in Alzheimer's disease (AD) preferentially afflicts the limbic and recently enlarged association cortices, causing a progression of mnemonic and cognitive deficits. Although genetic mouse models have helped reveal mechanisms underlying the rare, autosomal-dominant forms of AD, the etiology of the more common, sporadic form of AD remains unknown, and is challenging to study in mice due to their limited association cortex and lifespan. It is also difficult to study in human brains, as early-stage tau phosphorylation can degrade postmortem. In contrast, rhesus monkeys have extensive association cortices, are long-lived, and can undergo perfusion fixation to capture early-stage tau phosphorylation in situ. Most importantly, rhesus monkeys naturally develop amyloid plaques, neurofibrillary tangles comprised of hyperphosphorylated tau, synaptic loss, and cognitive deficits with advancing age, and thus can be used to identify the early molecular events that initiate and propel neuropathology in the aging association cortices. Studies to date suggest that the particular molecular signaling events needed for higher cognition-for example, high levels of calcium to maintain persistent neuronal firing- lead to tau phosphorylation and inflammation when dysregulated with advancing age. The expression of NMDAR-NR2B (GluN2B)-the subunit that fluxes high levels of calcium-increases over the cortical hierarchy and with the expansion of association cortex in primate evolution, consistent with patterns of tau pathology. In the rhesus monkey dorsolateral prefrontal cortex, spines contain NMDAR-NR2B and the molecular machinery to magnify internal calcium release near the synapse, as well as phosphodiesterases, mGluR3, and calbindin to regulate calcium signaling. Loss of regulation with inflammation and/or aging appears to be a key factor in initiating tau pathology. The vast expansion in the numbers of these synapses over primate evolution is consistent with the degree of tau pathology seen across species: marmoset < rhesus monkey < chimpanzee < human, culminating in the vast neurodegeneration seen in humans with AD.
Collapse
Affiliation(s)
- Amy F. T. Arnsten
- Department of NeuroscienceYale Medical SchoolNew HavenConnecticutUSA
| | - Dibyadeep Datta
- Department of NeuroscienceYale Medical SchoolNew HavenConnecticutUSA
| | - Todd M. Preuss
- Division of Neuropharmacology and Neurologic Diseases, Department of Pathology, Yerkes National Primate Research CenterEmory UniversityAtlantaGeorgiaUSA
| |
Collapse
|
30
|
Effects of maternal psychological stress during pregnancy on offspring brain development: Considering the role of inflammation and potential for preventive intervention. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2021; 7:461-470. [PMID: 34718150 PMCID: PMC9043032 DOI: 10.1016/j.bpsc.2021.10.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 10/04/2021] [Accepted: 10/18/2021] [Indexed: 11/22/2022]
Abstract
Heightened psychological stress during pregnancy has repeatedly been associated with increased risk for offspring development of behavior problems and psychiatric disorders. This review covers a rapidly growing body of research with the potential to advance a mechanistic understanding of these associations grounded in knowledge about maternal-placental-fetal stress biology and fetal brain development. Specifically, we highlight research employing magnetic resonance imaging to examine the infant brain soon after birth in relation to maternal psychological stress during pregnancy to increase capacity to identify specific alterations in brain structure and function and to differentiate between effects of pre- versus postnatal exposures. We then focus on heightened maternal inflammation during pregnancy as a mechanism through which maternal stress influences the developing fetal brain based on extensive preclinical literature and emerging research in humans. We place these findings in the context of recent work identifying psychotherapeutic interventions found to be effective for reducing psychological stress among pregnant individuals, which also show promise for reducing inflammation. We argue that a focus on inflammation, among other mechanistic pathways, has the potential to lead to a productive and necessary integration of research focused on the effects of maternal psychological stress on offspring brain development and prevention and intervention studies aimed at reducing maternal psychological stress during pregnancy. In addition to increasing capacity for common measurements and understanding potential mechanisms of action relevant to maternal mental health and fetal neurodevelopment, this focus can inform and broaden thinking about prevention and intervention strategies.
Collapse
|
31
|
Woo E, Sansing LH, Arnsten AFT, Datta D. Chronic Stress Weakens Connectivity in the Prefrontal Cortex: Architectural and Molecular Changes. CHRONIC STRESS 2021; 5:24705470211029254. [PMID: 34485797 PMCID: PMC8408896 DOI: 10.1177/24705470211029254] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 06/14/2021] [Indexed: 12/26/2022]
Abstract
Chronic exposure to uncontrollable stress causes loss of spines and dendrites in the prefrontal cortex (PFC), a recently evolved brain region that provides top-down regulation of thought, action, and emotion. PFC neurons generate top-down goals through recurrent excitatory connections on spines. This persistent firing is the foundation for higher cognition, including working memory, and abstract thought. However, exposure to acute uncontrollable stress drives high levels of catecholamine release in the PFC, which activates feedforward calcium-cAMP signaling pathways to open nearby potassium channels, rapidly weakening synaptic connectivity to reduce persistent firing. Chronic stress exposures can further exacerbate these signaling events leading to loss of spines and resulting in marked cognitive impairment. In this review, we discuss how stress signaling mechanisms can lead to spine loss, including changes to BDNF-mTORC1 signaling, calcium homeostasis, actin dynamics, and mitochondrial actions that engage glial removal of spines through inflammatory signaling. Stress signaling events may be amplified in PFC spines due to cAMP magnification of internal calcium release. As PFC dendritic spine loss is a feature of many cognitive disorders, understanding how stress affects the structure and function of the PFC will help to inform strategies for treatment and prevention.
Collapse
Affiliation(s)
- Elizabeth Woo
- Department of Neuroscience, Yale Medical School, New Haven, CT, USA.,Department of Neurology, Yale Medical School, New Haven, CT, USA
| | - Lauren H Sansing
- Department of Neurology, Yale Medical School, New Haven, CT, USA
| | - Amy F T Arnsten
- Department of Neuroscience, Yale Medical School, New Haven, CT, USA
| | - Dibyadeep Datta
- Department of Neuroscience, Yale Medical School, New Haven, CT, USA
| |
Collapse
|
32
|
Prieto-Villalobos J, Alvear TF, Liberona A, Lucero CM, Martínez-Araya CJ, Balmazabal J, Inostroza CA, Ramírez G, Gómez GI, Orellana JA. Astroglial Hemichannels and Pannexons: The Hidden Link between Maternal Inflammation and Neurological Disorders. Int J Mol Sci 2021; 22:ijms22179503. [PMID: 34502412 PMCID: PMC8430734 DOI: 10.3390/ijms22179503] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 08/30/2021] [Accepted: 08/30/2021] [Indexed: 12/11/2022] Open
Abstract
Maternal inflammation during pregnancy causes later-in-life alterations of the offspring’s brain structure and function. These abnormalities increase the risk of developing several psychiatric and neurological disorders, including schizophrenia, intellectual disability, bipolar disorder, autism spectrum disorder, microcephaly, and cerebral palsy. Here, we discuss how astrocytes might contribute to postnatal brain dysfunction following maternal inflammation, focusing on the signaling mediated by two families of plasma membrane channels: hemi-channels and pannexons. [Ca2+]i imbalance linked to the opening of astrocytic hemichannels and pannexons could disturb essential functions that sustain astrocytic survival and astrocyte-to-neuron support, including energy and redox homeostasis, uptake of K+ and glutamate, and the delivery of neurotrophic factors and energy-rich metabolites. Both phenomena could make neurons more susceptible to the harmful effect of prenatal inflammation and the experience of a second immune challenge during adulthood. On the other hand, maternal inflammation could cause excitotoxicity by producing the release of high amounts of gliotransmitters via astrocytic hemichannels/pannexons, eliciting further neuronal damage. Understanding how hemichannels and pannexons participate in maternal inflammation-induced brain abnormalities could be critical for developing pharmacological therapies against neurological disorders observed in the offspring.
Collapse
Affiliation(s)
- Juan Prieto-Villalobos
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (J.P.-V.); (T.F.A.); (A.L.); (C.J.M.-A.); (J.B.); (C.A.I.); (G.R.)
| | - Tanhia F. Alvear
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (J.P.-V.); (T.F.A.); (A.L.); (C.J.M.-A.); (J.B.); (C.A.I.); (G.R.)
| | - Andrés Liberona
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (J.P.-V.); (T.F.A.); (A.L.); (C.J.M.-A.); (J.B.); (C.A.I.); (G.R.)
| | - Claudia M. Lucero
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago 8910060, Chile; (C.M.L.); (G.I.G.)
| | - Claudio J. Martínez-Araya
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (J.P.-V.); (T.F.A.); (A.L.); (C.J.M.-A.); (J.B.); (C.A.I.); (G.R.)
| | - Javiera Balmazabal
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (J.P.-V.); (T.F.A.); (A.L.); (C.J.M.-A.); (J.B.); (C.A.I.); (G.R.)
| | - Carla A. Inostroza
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (J.P.-V.); (T.F.A.); (A.L.); (C.J.M.-A.); (J.B.); (C.A.I.); (G.R.)
| | - Gigliola Ramírez
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (J.P.-V.); (T.F.A.); (A.L.); (C.J.M.-A.); (J.B.); (C.A.I.); (G.R.)
| | - Gonzalo I. Gómez
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago 8910060, Chile; (C.M.L.); (G.I.G.)
| | - Juan A. Orellana
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (J.P.-V.); (T.F.A.); (A.L.); (C.J.M.-A.); (J.B.); (C.A.I.); (G.R.)
- Correspondence: ; Tel.: +56-23548105
| |
Collapse
|
33
|
Afzali M, Nilsson M, Palombo M, Jones DK. SPHERIOUSLY? The challenges of estimating sphere radius non-invasively in the human brain from diffusion MRI. Neuroimage 2021; 237:118183. [PMID: 34020013 PMCID: PMC8285594 DOI: 10.1016/j.neuroimage.2021.118183] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 04/25/2021] [Accepted: 05/16/2021] [Indexed: 11/16/2022] Open
Abstract
The Soma and Neurite Density Imaging (SANDI) three-compartment model was recently proposed to disentangle cylindrical and spherical geometries, attributed to neurite and soma compartments, respectively, in brain tissue. There are some recent advances in diffusion-weighted MRI signal encoding and analysis (including the use of multiple so-called 'b-tensor' encodings and analysing the signal in the frequency-domain) that have not yet been applied in the context of SANDI. In this work, using: (i) ultra-strong gradients; (ii) a combination of linear, planar, and spherical b-tensor encodings; and (iii) analysing the signal in the frequency domain, three main challenges to robust estimation of sphere size were identified: First, the Rician noise floor in magnitude-reconstructed data biases estimates of sphere properties in a non-uniform fashion. It may cause overestimation or underestimation of the spherical compartment size and density. This can be partly ameliorated by accounting for the noise floor in the estimation routine. Second, even when using the strongest diffusion-encoding gradient strengths available for human MRI, there is an empirical lower bound on the spherical signal fraction and radius that can be detected and estimated robustly. For the experimental setup used here, the lower bound on the sphere signal fraction was approximately 10%. We employed two different ways of establishing the lower bound for spherical radius estimates in white matter. The first, examining power-law relationships between the DW-signal and diffusion weighting in empirical data, yielded a lower bound of 7μm, while the second, pure Monte Carlo simulations, yielded a lower limit of 3μm and in this low radii domain, there is little differentiation in signal attenuation. Third, if there is sensitivity to the transverse intra-cellular diffusivity in cylindrical structures, e.g., axons and cellular projections, then trying to disentangle two diffusion-time-dependencies using one experimental parameter (i.e., change in frequency-content of the encoding waveform) makes spherical radii estimates particularly challenging. We conclude that due to the aforementioned challenges spherical radii estimates may be biased when the corresponding sphere signal fraction is low, which must be considered.
Collapse
Affiliation(s)
- Maryam Afzali
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, United Kingdom.
| | - Markus Nilsson
- Clinical Sciences Lund, Radiology, Lund University, Lund, Sweden.
| | - Marco Palombo
- Centre for Medical Image Computing, Department of Computer Science, University College London, London, United Kingdom.
| | - Derek K Jones
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, United Kingdom.
| |
Collapse
|
34
|
Réus GZ, Giridharan VV, de Moura AB, Borba LA, Botelho MEM, Behenck JP, Generoso JS, Selvaraj S, Bhatti G, Barichello T, Quevedo J. The impact of early life stress and immune challenge on behavior and glia cells alteration in late adolescent rats. Int J Dev Neurosci 2021; 81:407-415. [PMID: 33788296 DOI: 10.1002/jdn.10108] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/08/2021] [Accepted: 03/25/2021] [Indexed: 01/06/2023] Open
Abstract
Maternal deprivation (MD) is known to be related to long-term changes that could influence the onset of psychiatric disorders. Studies have demonstrated that early life stress makes the cells in the brain more susceptible to subsequent stressors. To test it, we used an animal model of MD conducted from postnatal day (PND) 1 to 10. Deprived and non-deprived rats (control) were randomized to receive or not lipopolysaccharide (LPS) at 5 mg/kg on PND 50. The behavior and glial cells activation were evaluated in all groups from 51 to 53 PND. There was an increase in the immobility time in the MD and MD+LPS groups. The spontaneous locomotor activity was not changed between groups. We found elevated ionized calcium-binding adapter molecule 1 (Iba-1)-positive cells levels in the control+LPS and MD+LPS groups. In the MD+LPS group, it was found an increase in Iba-positive cells compared to the MD+sal group. The glial fibrillary acidic protein (GFAP)-positive cells were also increased in the MD+LPS, compared to control+sal, control+LPS, and MD+sal groups. Immune challenge by LPS in late adolescence, which was subjected to MD, did not influence the depressive-like behavior but exerted a pronounced effect in the microglial activation and astrocyte atrophy.
Collapse
Affiliation(s)
- Gislaine Z Réus
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Vijayasree V Giridharan
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Airam B de Moura
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Laura A Borba
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Maria Eduarda M Botelho
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - João Paulo Behenck
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Jaqueline S Generoso
- Laboratory of Experimental Microbiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Sudhakar Selvaraj
- Louis Faillace, MD, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Gursimrat Bhatti
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Tatiana Barichello
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Laboratory of Experimental Microbiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - João Quevedo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Neuroscience Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA
| |
Collapse
|
35
|
Arnsten AFT, Datta D, Wang M. The genie in the bottle-magnified calcium signaling in dorsolateral prefrontal cortex. Mol Psychiatry 2021; 26:3684-3700. [PMID: 33319854 PMCID: PMC8203737 DOI: 10.1038/s41380-020-00973-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/20/2020] [Accepted: 11/26/2020] [Indexed: 02/07/2023]
Abstract
Neurons in the association cortices are particularly vulnerable in cognitive disorders such as schizophrenia and Alzheimer's disease, while those in primary visual cortex remain relatively resilient. This review proposes that the special molecular mechanisms needed for higher cognitive operations confer vulnerability to dysfunction, atrophy, and neurodegeneration when regulation is lost due to genetic and/or environmental insults. Accumulating data suggest that higher cortical circuits rely on magnified levels of calcium (from NMDAR, calcium channels, and/or internal release from the smooth endoplasmic reticulum) near the postsynaptic density to promote the persistent firing needed to maintain, manipulate, and store information without "bottom-up" sensory stimulation. For example, dendritic spines in the primate dorsolateral prefrontal cortex (dlPFC) express the molecular machinery for feedforward, cAMP-PKA-calcium signaling. PKA can drive internal calcium release and promote calcium flow through NMDAR and calcium channels, while in turn, calcium activates adenylyl cyclases to produce more cAMP-PKA signaling. Excessive levels of cAMP-calcium signaling can have a number of detrimental effects: for example, opening nearby K+ channels to weaken synaptic efficacy and reduce neuronal firing, and over a longer timeframe, driving calcium overload of mitochondria to induce inflammation and dendritic atrophy. Thus, calcium-cAMP signaling must be tightly regulated, e.g., by agents that catabolize cAMP or inhibit its production (PDE4, mGluR3), and by proteins that bind calcium in the cytosol (calbindin). Many genetic or inflammatory insults early in life weaken the regulation of calcium-cAMP signaling and are associated with increased risk of schizophrenia (e.g., GRM3). Age-related loss of regulatory proteins which result in elevated calcium-cAMP signaling over a long lifespan can additionally drive tau phosphorylation, amyloid pathology, and neurodegeneration, especially when protective calcium binding proteins are lost from the cytosol. Thus, the "genie" we need for our remarkable cognitive abilities may make us vulnerable to cognitive disorders when we lose essential regulation.
Collapse
Affiliation(s)
- Amy F T Arnsten
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, 06510, USA.
| | - Dibyadeep Datta
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Min Wang
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, 06510, USA
| |
Collapse
|
36
|
da Conceição Pereira S, Manhães-de-Castro R, Visco DB, de Albuquerque GL, da Silva Calado CMS, da Silva Souza V, Toscano AE. Locomotion is impacted differently according to the perinatal brain injury model: Meta-analysis of preclinical studies with implications for cerebral palsy. J Neurosci Methods 2021; 360:109250. [PMID: 34116077 DOI: 10.1016/j.jneumeth.2021.109250] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/03/2021] [Accepted: 06/05/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Different approaches to reproduce cerebral palsy (CP) in animals, contribute to the knowledge of the pathophysiological mechanism of this disease and provide a basis for the development of intervention strategies. Locomotion and coordination are the main cause of disability in CP, however, few studies highlight the quantitative differences of CP models, on locomotion parameters, considering the methodologies to cause brain lesions in the perinatal period. METHODS Studies with cerebral palsy animal models that assess locomotion parameters were systematically retrieved from Medline/PubMed, SCOPUS, LILACS, and Web of Science. Methodological evaluation of included studies and quantitative assessment of locomotion parameters were performed after eligibility screening. RESULTS CP models were induced by hypoxia-ischemia (HI), Prenatal ischemia (PI), lipopolysaccharide inflammation (LPS), intraventricular haemorrhage (IVH), anoxia (A), sensorimotor restriction (SR), and a combination of different models. Overall, 63 studies included in qualitative synthesis showed a moderate quality of evidence. 16 studies were included in the quantitative meta-analysis. Significant reduction was observed in models that combined LPS with HI related to distance traveled (SMD -7.24 95 % CI [-8.98, -5.51], Z = 1.18, p < 0.00001) and LPS with HI or anoxia with sensory-motor restriction (SMD -6.01, 95 % CI [-7.67, -4.35], Z = 7.11), or IVH (SMD -4.91, 95 % CI [-5.84, -3.98], Z = 10.31, p < 0.00001) related to motor coordination. CONCLUSION The combination of different approaches to reproduce CP in animals causes greater deficits in locomotion and motor coordination from the early stages of life to adulthood. These findings contribute to methodological refinement, reduction, and replacement in animal experimentation, favoring translational purposes.
Collapse
Affiliation(s)
- Sabrina da Conceição Pereira
- Posgraduate Program in Neuropsychiatry and Behavior Sciences, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Raul Manhães-de-Castro
- Posgraduate Program in Neuropsychiatry and Behavior Sciences, Federal University of Pernambuco, Recife, Pernambuco, Brazil; Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Pernambuco, Brazil; Postgraduate Program in Nutrition, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Diego Bulcão Visco
- Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Pernambuco, Brazil; Postgraduate Program in Nutrition, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | | | | | - Vanessa da Silva Souza
- Posgraduate Program in Neuropsychiatry and Behavior Sciences, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Ana Elisa Toscano
- Posgraduate Program in Neuropsychiatry and Behavior Sciences, Federal University of Pernambuco, Recife, Pernambuco, Brazil; Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Pernambuco, Brazil; Postgraduate Program in Nutrition, Federal University of Pernambuco, Recife, Pernambuco, Brazil; Department of Nursing, CAV, Federal University of Pernambuco, Vitória de Santo Antão, Pernambuco, Brazil.
| |
Collapse
|
37
|
Huizing MJ, Borges-Luján M, Cavallaro G, González-Luis GE, Raffaeli G, Bas-Suárez P, Bakker JA, Moonen RM, Villamor E. Plasma Amino Acid Concentrations at Birth and Patent Ductus Arteriosus in Very and Extremely Preterm Infants. Front Pediatr 2021; 9:647018. [PMID: 33643980 PMCID: PMC7905031 DOI: 10.3389/fped.2021.647018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 01/20/2021] [Indexed: 11/17/2022] Open
Abstract
Background: Amino acids are increasingly recognized as bioactive molecules in numerous physiological and pathophysiological pathways. The non-essential amino acid glutamate is vasoactive in the rat ductus arteriosus (DA) and a decrease in its levels within the 1st days of life has been associated with the presence of patent DA (PDA) in extremely preterm infants. However, these findings have not been confirmed in other studies. Objective: To investigate the possible association between amino acid concentrations in the 1st day of life and the presence of PDA in a cohort of 121 newborns with gestational age (GA) below 30 weeks and birth weight (BW) below 1,500 g. Methods: Plasma samples were collected 6-12 h after birth and amino acid concentrations were determined by tandem mass spectrometry. Besides PDA, we analyzed the potential association of amino acid concentrations with infant sex, small for GA (SGA, defined as BW < third percentile), antenatal corticosteroids, chorioamnionitis, and preeclampsia. Group differences were analyzed by ANOVA adjusted for GA and BW. A Bonferroni significance threshold of P < 0.0024 was used to correct for multiple testing. Results: PDA was found in 48 of the 121 infants examined. We observed higher mean levels of glutamate in infants with PDA (147.0 μmol/L, SD 84.0) as compared with those without (106.7 μmol/L, SD 49.1, P = 0.0006). None of the other amino acid concentrations in the PDA group reached the level of statistical significance that was pre-set to correct for multiple comparisons. Glutamate levels were not significantly affected by infant sex, being SGA, or by exposure to antenatal corticosteroids, clinical chorioamnionitis, or preeclampsia. Conclusion: Our study not only does not confirm the previous findings of low glutamate levels in preterm infants with PDA, but we have even found elevated glutamate concentrations associated with PDA. Nevertheless, despite the high statistical significance, the difference in glutamate levels may lack clinical significance or may be an epiphenomenon associated with the particular clinical condition of infants with PDA.
Collapse
Affiliation(s)
- Maurice J Huizing
- Department of Pediatrics, Maastricht University Medical Centre (MUMC+), School for Oncology and Developmental Biology (GROW), Maastricht, Netherlands
| | - Moreyba Borges-Luján
- Department of Neonatology, Complejo Hospitalario Universitario Insular Materno-Infantil (CHUIMI) de Canarias, Las Palmas de Gran Canaria, Spain
| | - Giacomo Cavallaro
- Neonatal Intensive Care Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Gema E González-Luis
- Department of Neonatology, Complejo Hospitalario Universitario Insular Materno-Infantil (CHUIMI) de Canarias, Las Palmas de Gran Canaria, Spain
| | - Genny Raffaeli
- Neonatal Intensive Care Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Pilar Bas-Suárez
- Department of Pediatrics, Hospital Vithas Santa Catalina, Las Palmas de Gran Canaria, Spain
| | - Jaap A Bakker
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Rob M Moonen
- Department of Pediatrics, Zuyderland Medical Center, Heerlen, Netherlands
| | - Eduardo Villamor
- Department of Pediatrics, Maastricht University Medical Centre (MUMC+), School for Oncology and Developmental Biology (GROW), Maastricht, Netherlands
| |
Collapse
|
38
|
Arnsten AFT, Wang M. The Evolutionary Expansion of mGluR3-NAAG-GCPII Signaling: Relevance to Human Intelligence and Cognitive Disorders. Am J Psychiatry 2020; 177:1103-1106. [PMID: 33256450 DOI: 10.1176/appi.ajp.2020.20101458] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Affiliation(s)
- Amy F T Arnsten
- Department of Neuroscience, Yale University School of Medicine, New Haven Conn. (Arnsten, Wang)
| | - Min Wang
- Department of Neuroscience, Yale University School of Medicine, New Haven Conn. (Arnsten, Wang)
| |
Collapse
|
39
|
Arteaga Cabeza O, Zhang Z, Smith Khoury E, Sheldon RA, Sharma A, Zhang F, Slusher BS, Kannan RM, Kannan S, Ferriero DM. Neuroprotective effects of a dendrimer-based glutamate carboxypeptidase inhibitor on superoxide dismutase transgenic mice after neonatal hypoxic-ischemic brain injury. Neurobiol Dis 2020; 148:105201. [PMID: 33271328 PMCID: PMC8351403 DOI: 10.1016/j.nbd.2020.105201] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/28/2020] [Accepted: 11/23/2020] [Indexed: 01/05/2023] Open
Abstract
The result of a deprivation of oxygen and glucose to the brain, hypoxic-ischemic encephalopathy (HIE), remains the most common cause of death and disability in human neonates globally and is mediated by glutamate toxicity and inflammation. We have previously shown that the enzyme glutamate carboxypeptidase (GCPII) is overexpressed in activated microglia in the presence of inflammation in fetal/newborn rabbit brain. We assessed the therapeutic utility of a GCPII enzyme inhibitor called 2-(3-Mercaptopropyl) pentanedioic acid (2MPPA) attached to a dendrimer (D-2MPPA), in order to target activated microglia in an experimental neonatal hypoxia-ischemia (HI) model using superoxide dismutase transgenic (SOD) mice that are often more injured after hypoxia-ischemia than wildtype animals. SOD overexpressing and wild type (WT) mice underwent permanent ligation of the left common carotid artery followed by 50 min of asphyxiation (10% O2) to induce HI injury on postnatal day 9 (P9). Cy5-labeled dendrimers were administered to the mice at 6 h, 24 h or 72 h after HI and brains were evaluated by immunofluorescence analysis 24 h after the injection to visualize microglial localization and uptake over time. Expression of GCPII enzyme was analyzed in microglia 24 h after the HI injury. The expression of pro- and anti-inflammatory cytokines were analyzed 24 h and 72 h post-HI. Brain damage was analyzed histologically 7 days post-HI in the three randomly assigned groups: control (C); hypoxic-ischemic (HI); and HI mice who received a single dose of D-2MPPA 6 h post-HI (HI+D-2MPPA). First, we found that GCPII was overexpressed in activated microglia 24 h after HI in the SOD overexpressing mice. Also, there was an increase in microglial activation 24 h after HI in the ipsilateral hippocampus which was most visible in the SOD+HI group. Dendrimers were mostly taken up by microglia by 24 h post-HI; uptake was more prominent in the SOD+HI mice than in the WT+HI. The inflammatory profile showed significant increase in expression of KC/GRO following injury in SOD mice compared to WT at 24 and 72 h. A greater and significant decrease in KC/GRO was seen in the SOD mice following treatment with D-2MPPA. Seven days after HI, D-2MPPA treatment decreased brain injury in the SOD+HI group, but not in WT+HI. This reduced damage was mainly seen in hippocampus and cortex. Our data indicate that the best time point to administer D-2MPPA is 6 h post-HI in order to suppress the expression of GCPII by 24 h after the damage since dendrimer localization in microglia is seen as early as 6 h with the peak of GCPII upregulation in activated microglia seen at 24 h post-HI. Ultimately, treatment with D-2MPPA at 6 h post-HI leads to a decrease in inflammatory profiles by 24 h and reduction in brain injury in the SOD overexpressing mice.
Collapse
Affiliation(s)
- O Arteaga Cabeza
- Departments of Pediatrics, University of California San Francisco, San Francisco, CA 94158, USA
| | - Z Zhang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - E Smith Khoury
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - R A Sheldon
- Departments of Pediatrics, University of California San Francisco, San Francisco, CA 94158, USA; Departments of Newborn Brain Research Institute, University of California San Francisco, San Francisco, CA 94158, USA
| | - A Sharma
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - F Zhang
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - B S Slusher
- Department of Neurology, Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - R M Kannan
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - S Kannan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - D M Ferriero
- Departments of Pediatrics, University of California San Francisco, San Francisco, CA 94158, USA; Departments of Neurology, University of California San Francisco, San Francisco, CA 94158, USA; Departments of Newborn Brain Research Institute, University of California San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
40
|
Zhang Z, Lin YA, Kim SY, Su L, Liu J, Kannan RM, Kannan S. Systemic dendrimer-drug nanomedicines for long-term treatment of mild-moderate cerebral palsy in a rabbit model. J Neuroinflammation 2020; 17:319. [PMID: 33100217 PMCID: PMC7586697 DOI: 10.1186/s12974-020-01984-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 10/05/2020] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Neuroinflammation mediated by microglia plays a central role in the pathogenesis of perinatal/neonatal brain injury, including cerebral palsy (CP). Therapeutics mitigating neuroinflammation potentially provide an effective strategy to slow the disease progression and rescue normal brain development. Building on our prior results which showed that a generation-4 hydroxyl poly(amidoamine) (PAMAM) dendrimer could deliver drugs specifically to activated glia from systemic circulation, we evaluated the sustained efficacy of a generation-6 (G6) hydroxyl-terminated PAMAM dendrimer that showed a longer blood circulation time and increased brain accumulation. N-acetyl-L-cysteine (NAC), an antioxidant and anti-inflammatory agent that has high plasma protein binding properties and poor brain penetration, was conjugated to G6-PAMAM dendrimer-NAC (G6D-NAC). The efficacy of microglia-targeted G6D-NAC conjugate was evaluated in a clinically relevant rabbit model of CP, with a mild/moderate CP phenotype to provide a longer survival of untreated CP kits, enabling the assessment of sustained efficacy over 15 days of life. METHODS G6D-NAC was conjugated and characterized. Cytotoxicity and anti-inflammatory assays were performed in BV-2 microglial cells. The efficacy of G6D-NAC was evaluated in a rabbit model of CP. CP kits were randomly divided into 5 groups on postnatal day 1 (PND1) and received an intravenous injection of a single dose of PBS, or G6D-NAC (2 or 5 mg/kg), or NAC (2 or 5 mg/kg). Neurobehavioral tests, microglia morphology, and neuroinflammation were evaluated at postnatal day 5 (PND5) and day 15 (PND15). RESULTS A single dose of systemic 'long circulating' G6D-NAC showed a significant penetration across the impaired blood-brain-barrier (BBB), delivered NAC specifically to activated microglia, and significantly reduced microglia-mediated neuroinflammation in both the cortex and cerebellum white matter areas. Moreover, G6D-NAC treatment significantly improved neonatal rabbit survival rate and rescued motor function to nearly healthy control levels at least up to 15 days after birth (PND15), while CP kits treated with free NAC died before PND9. CONCLUSIONS Targeted delivery of therapeutics to activated microglia in neonatal brain injury can ameliorate pro-inflammatory microglial responses to injury, promote survival rate, and improve neurological outcomes that can be sustained for a long period. Appropriate manipulation of activated microglia enabled by G6D-NAC can impact the injury significantly beyond inflammation.
Collapse
Affiliation(s)
- Zhi Zhang
- Department of Anesthesiology and Critical Care, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Present address: Department of Natural Sciences, University of Michigan-Dearborn, Dearborn, MI, USA
| | - Yi-An Lin
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins School of Medicine, 400 North Broadway, Baltimore, MD, 21287, USA
| | - Soo-Young Kim
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins School of Medicine, 400 North Broadway, Baltimore, MD, 21287, USA
| | - Lilly Su
- Department of Anesthesiology and Critical Care, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jinhuan Liu
- Department of Anesthesiology and Critical Care, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Rangaramanujam M Kannan
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins School of Medicine, 400 North Broadway, Baltimore, MD, 21287, USA.
| | - Sujatha Kannan
- Department of Anesthesiology and Critical Care, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Department of Anesthesiology and Critical Care Medicine, Charlotte Bloomberg Children's Center 6318D, 1800 Orleans Street, Baltimore, MD, 21287, USA.
| |
Collapse
|
41
|
Bratek E, Ziembowicz A, Salinska E. N-Acetylaspartylglutamate (NAAG) Pretreatment Reduces Hypoxic-Ischemic Brain Damage and Oxidative Stress in Neonatal Rats. Antioxidants (Basel) 2020; 9:antiox9090877. [PMID: 32957477 PMCID: PMC7555246 DOI: 10.3390/antiox9090877] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/08/2020] [Accepted: 09/14/2020] [Indexed: 12/15/2022] Open
Abstract
N-acetylaspartylglutamate (NAAG), the most abundant peptide transmitter in the mammalian nervous system, activates mGluR3 at presynaptic sites, inhibiting the release of glutamate, and acts on mGluR3 on astrocytes, stimulating the release of neuroprotective growth factors (TGF-β). NAAG can also affect N-methyl-d-aspartate (NMDA) receptors in both synaptic and extrasynaptic regions. NAAG reduces neurodegeneration in a neonatal rat model of hypoxia-ischemia (HI), although the exact mechanism is not fully recognized. In the present study, the effect of NAAG application 24 or 1 h before experimental birth asphyxia on oxidative stress markers and the potential mechanisms of neuroprotection on 7-day old rats was investigated. The intraperitoneal application of NAAG at either time point before HI significantly reduced the weight deficit of the ischemic brain hemisphere, radical oxygen species (ROS) content and activity of antioxidant enzymes, and increased the concentration of reduced glutathione (GSH). No additional increase in the TGF-β concentration was observed after NAAG application. The fast metabolism of NAAG and the decrease in TGF-β concentration that resulted from NAAG pretreatment, performed up to 24 h before HI, excluded the involvement mGluR3 in neuroprotection. The observed effect may be explained by the activation of NMDA receptors induced by NAAG pretreatment 24 h before HI. Inhibition of the NAAG effect by memantine supports this conclusion. NAAG preconditioning 1 h before HI results in a mixture of mGluR3 and NMDA receptor activation. Preconditioning with NAAG induces the antioxidative defense system triggered by mild excitotoxicity in neurons. Moreover, this response to NAAG pretreatment is consistent with the commonly accepted mechanism of preconditioning. However, this theory requires further investigation.
Collapse
|
42
|
Yap V, Perlman JM. Mechanisms of brain injury in newborn infants associated with the fetal inflammatory response syndrome. Semin Fetal Neonatal Med 2020; 25:101110. [PMID: 32303463 DOI: 10.1016/j.siny.2020.101110] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The fetal inflammatory response syndrome (FIRS) is characterized by umbilical cord inflammation and elevated fetal pro-inflammatory cytokines. Surviving neonates, especially very preterm infants, have increased rates of neonatal morbidity including neurodevelopmental impairment. The mechanism of brain injury in FIRS is complex and may involve "multiple hits." Exposure to in utero inflammation initiates a cascade of the fetal immune response, where pro-inflammatory cytokines can cause direct injury to oligodendrocytes and neurons. Activation of microglia results in further injury to vulnerable pre-myelinating oligodendrocytes and influences the integrity of the fetal and newborn's blood-brain barrier, resulting in further exposure of the brain to developmental insults. Newborns exposed to FIRS are frequently exposed to additional perinatal and postnatal insults that can result in further brain injury. Future directions should include evaluations for new therapeutic interventions aimed at reducing brain injury by dampening FIRS, inhibition of microglial activation, and regeneration of immature oligodendrocytes.
Collapse
Affiliation(s)
- Vivien Yap
- Weill Cornell Medicine - New York Presbyterian Hospital, 525 East 68th Street, Suite N-506, New York, NY, 10065, United States.
| | - Jeffrey M Perlman
- Weill Cornell Medicine - New York Presbyterian Hospital, 525 East 68th Street, Suite N-506, New York, NY, 10065, United States
| |
Collapse
|
43
|
Zhang Z, Ishrat S, O'Bryan M, Klein B, Saraswati M, Robertson C, Kannan S. Pediatric Traumatic Brain Injury Causes Long-Term Deficits in Adult Hippocampal Neurogenesis and Cognition. J Neurotrauma 2020; 37:1656-1667. [DOI: 10.1089/neu.2019.6894] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Zhi Zhang
- Department of Natural Sciences, University of Michigan-Dearborn, Dearborn, Michigan, USA
| | - Samiha Ishrat
- Department of Natural Sciences, University of Michigan-Dearborn, Dearborn, Michigan, USA
| | - Megan O'Bryan
- Department of Natural Sciences, University of Michigan-Dearborn, Dearborn, Michigan, USA
| | - Brandon Klein
- Department of Natural Sciences, University of Michigan-Dearborn, Dearborn, Michigan, USA
| | - Manda Saraswati
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Courtney Robertson
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Sujatha Kannan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
44
|
Khoury ES, Sharma A, Ramireddy RR, Thomas AG, Alt J, Fowler A, Rais R, Tsukamoto T, Blue ME, Slusher B, Kannan S, Kannan RM. Dendrimer-conjugated glutaminase inhibitor selectively targets microglial glutaminase in a mouse model of Rett syndrome. Am J Cancer Res 2020; 10:5736-5748. [PMID: 32483415 PMCID: PMC7254984 DOI: 10.7150/thno.41714] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 04/14/2020] [Indexed: 12/14/2022] Open
Abstract
Background: Elevated glutamate production and release from glial cells is a common feature of many CNS disorders. Inhibitors of glutaminase (GLS), the enzyme responsible for converting glutamine to glutamate have been developed to target glutamate overproduction. However, many GLS inhibitors have poor aqueous solubility, are unable to cross the blood brain barrier, or demonstrate significant toxicity when given systemically, precluding translation. Enhanced aqueous solubility and systemic therapy targeted to activated glia may address this challenge. Here we examine the impact of microglial-targeted GLS inhibition in a mouse model of Rett syndrome (RTT), a developmental disorder with no viable therapies, manifesting profound central nervous system effects, in which elevated glutamatergic tone, upregulation of microglial GLS, oxidative stress and neuroimmune dysregulation are key features. Methods: To enable this, we conjugated a potent glutaminase inhibitor, N-(5-{2-[2-(5-amino-[1,3,4]thiadiazol-2-yl)-ethylsulfanyl]-ethyl}-[1,3,4]thiadiazol-2-yl)-2-phenyl-acetamide (JHU29) to a generation 4 hydroxyl PAMAM dendrimer (D-JHU29). We then examined the effect of D-JHU29 in organotypic slice culture on glutamate release. We also examined GLS activity in microglial and non-microglial cells, and neurobehavioral phenotype after systemic administration of D-JHU29 in a mouse model of RTT. Results: We report successful conjugation of JHU29 to dendrimer resulting in enhanced water solubility compared to free JHU29. D-JHU29 reduced the excessive glutamate release observed in tissue culture slices in a clinically relevant Mecp2-knockout (KO) RTT mouse. Microglia isolated from Mecp2-KO mice demonstrated upregulation of GLS activity that normalized to wild-type levels following systemic treatment with D-JHU29. Neurobehavioral assessments in D-JHU29 treated Mecp2-KO mice revealed selective improvements in mobility. Conclusion: These findings demonstrate that glutaminase inhibitors conjugated to dendrimers are a viable mechanism to selectively inhibit microglial GLS to reduce glutamate production and improve mobility in a mouse model of RTT, with broader implications for selectively targeting this pathway in other neurodegenerative disorders.
Collapse
|
45
|
Bae H, Lee JY, Song G, Lim W. Function of CCL5 in maternal-fetal interface of pig during early pregnancy. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 103:103503. [PMID: 31563460 DOI: 10.1016/j.dci.2019.103503] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/25/2019] [Accepted: 09/26/2019] [Indexed: 06/10/2023]
Abstract
Chemokines refer to chemoattractant cytokines, which have crucial functions in inflammation and immune responses in multiple cellular processes. In the present study, we described the potential role of porcine CCL5 in embryo implantation and fetal-maternal environment during early pregnancy. We first carried out phylogenetic analysis of porcine CCL5, and analyzed the cell specific localization of CCL5 and its receptor CCR3 in a kinetic approach within porcine estrous cycles and early gestation stage. In addition, CCL5 stimulated porcine uterine luminal epithelial (pLE) and porcine trophectoderm (pTr) cell proliferations, and cell cycle progressions via AKT and MAPK intracellular signaling tractions. Furthermore, CCL5 attenuated tunicamycin-induced endoplasmic reticulum (ER) stress signaling, and lipopolysaccharides-triggered inflammatory responses in pLE and pTr cells. Taken together, our study showed that CCL5 is involved in the placental development or promotes the placental development.
Collapse
Affiliation(s)
- Hyocheol Bae
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Jin-Young Lee
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Gwonhwa Song
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea.
| | - Whasun Lim
- Department of Food and Nutrition, Kookmin University, Seoul, 02707, Republic of Korea.
| |
Collapse
|
46
|
Jia Y, Chen Y, Geng K, Cheng Y, Li Y, Qiu J, Huang H, Wang R, Zhang Y, Wu R. Glutamate Chemical Exchange Saturation Transfer (GluCEST) Magnetic Resonance Imaging in Pre-clinical and Clinical Applications for Encephalitis. Front Neurosci 2020; 14:750. [PMID: 32848546 PMCID: PMC7399024 DOI: 10.3389/fnins.2020.00750] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 06/25/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Encephalitis is a common central nervous system inflammatory disease that seriously endangers human health owing to the lack of effective diagnostic methods, which leads to a high rate of misdiagnosis and mortality. Glutamate is implicated closely in microglial activation, and activated microglia are key players in encephalitis. Hence, using glutamate chemical exchange saturation transfer (GluCEST) imaging for the early diagnosis of encephalitis holds promise. METHODS The sensitivity of GluCEST imaging with different concentrations of glutamate and other major metabolites in the brain was validated in phantoms. Twenty-seven Sprague-Dawley (SD) rats with encephalitis induced by Staphylococcus aureus infection were used for preclinical research of GluCEST imaging in a 7.0-Tesla scanner. For the clinical study, six patients with encephalitis, six patients with lacunar infarction, and six healthy volunteers underwent GluCEST imaging in a 3.0-Tesla scanner. RESULTS The number of amine protons on glutamate that had a chemical shift of 3.0 ppm away from bulk water and the signal intensity of GluCEST were concentration-dependent. Under physiological conditions, glutamate is the main contributor to the GluCEST signal. Compared with normal tissue, in both rats and patients with encephalitis, the encephalitis areas demonstrated a hyper-intense GluCEST signal, while the lacunar infarction had a decreased GluCEST signal intensity. After intravenous immunoglobulin therapy, patients with encephalitis lesions showed a decrease in GluCEST signal, and the results were significantly different from the pre-treatment signal (1.34 ± 0.31 vs 5.0 ± 0.27%, respectively; p = 0.000). CONCLUSION Glutamate plays a role in encephalitis, and the GluCEST imaging signal has potential as an in vivo imaging biomarker for the early diagnosis of encephalitis. GluCEST will provide new insight into encephalitis and help improve the differential diagnosis of brain disorders.
Collapse
Affiliation(s)
- Yanlong Jia
- Department of Radiology, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Yanzi Chen
- Department of Radiology, Affiliated Longhua People’s Hospital, Southern Medical University, Shenzhen, China
| | - Kuan Geng
- Department of Radiology, The First People’s Hospital of Honghe Prefecture, Mengzi, China
| | - Yan Cheng
- Department of Radiology, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Yan Li
- Department of Radiology, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Jinming Qiu
- Department of Radiology, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Huaidong Huang
- Department of Radiology, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Runrun Wang
- Department of Radiology, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Yunping Zhang
- Department of Nuclear Medicine, Shenzhen Luohu District People’s Hospital, Shenzhen, China
- *Correspondence: Yunping Zhang,
| | - Renhua Wu
- Department of Radiology, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
- Renhua Wu,
| |
Collapse
|
47
|
Dunn GA, Nigg JT, Sullivan EL. Neuroinflammation as a risk factor for attention deficit hyperactivity disorder. Pharmacol Biochem Behav 2019; 182:22-34. [PMID: 31103523 PMCID: PMC6855401 DOI: 10.1016/j.pbb.2019.05.005] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 05/08/2019] [Accepted: 05/14/2019] [Indexed: 01/08/2023]
Abstract
Attention Deficit Hyperactivity Disorder (ADHD) is a persistent, and impairing pediatric-onset neurodevelopmental condition. Its high prevalence, and recurrent controversy over its widespread identification and treatment, drive strong interest in its etiology and mechanisms. Emerging evidence for a role for neuroinflammation in ADHD pathophysiology is of great interest. This evidence includes 1) the above-chance comorbidity of ADHD with inflammatory and autoimmune disorders, 2) initial studies indicating an association with ADHD and increased serum cytokines, 3) preliminary evidence from genetic studies demonstrating associations between polymorphisms in genes associated with inflammatory pathways and ADHD, 4) emerging evidence that early life exposure to environmental factors may increase risk for ADHD via an inflammatory mechanism, and 5) mechanistic evidence from animal models of maternal immune activation documenting behavioral and neural outcomes consistent with ADHD. Prenatal exposure to inflammation is associated with changes in offspring brain development including reductions in cortical gray matter volume and the volume of certain cortical areas -parallel to observations associated with ADHD. Alterations in neurotransmitter systems, including the dopaminergic, serotonergic and glutamatergic systems, are observed in ADHD populations. Animal models provide strong evidence that development and function of these neurotransmitters systems are sensitive to exposure to in utero inflammation. In summary, accumulating evidence from human studies and animal models, while still incomplete, support a potential role for neuroinflammation in the pathophysiology of ADHD. Confirmation of this association and the underlying mechanisms have become valuable targets for research. If confirmed, such a picture may be important in opening new intervention routes.
Collapse
Affiliation(s)
| | - Joel T Nigg
- Oregon Health and Science University, United States of America
| | - Elinor L Sullivan
- University of Oregon, United States of America; Oregon Health and Science University, United States of America; Oregon National Primate Research Center, United States of America.
| |
Collapse
|
48
|
Meng G, Zhou X, Wang M, Zhou L, Wang Z, Wang M, Deng J, Wang Y, Zhou Z, Zhang Y, Lai Y, Zhang Q, Yang X, Yu L, Jiang H. Gut microbe-derived metabolite trimethylamine N-oxide activates the cardiac autonomic nervous system and facilitates ischemia-induced ventricular arrhythmia via two different pathways. EBioMedicine 2019; 44:656-664. [PMID: 30954457 PMCID: PMC6603492 DOI: 10.1016/j.ebiom.2019.03.066] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 03/20/2019] [Accepted: 03/21/2019] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND We previously demonstrated the gut microbes-derived metabolite trimethylamine N-oxide (TMAO) could activate the atrial autonomic ganglion plexus and promote atrial arrhythmia. The cardiac sympathetic nervous system (CSNS) play important roles in modulating ventricular arrhythmia (VA). METHODS Part 1: To test whether TMAO can directly activate the CSNS, we performed local injection of TMAO into the left stellate ganglion (LSG). Part 2: To test whether TMAO can indirectly activate the CSNS through the central nervous system, we performed intravenous injection of TMAO. Ventricular electrophysiology and LSG function and neural activity were measured before and after TMAO administration. Then, the left anterior descending coronary artery was ligated, and electrocardiograms were recorded for 1 h. At the end of the experiment, LSG and paraventricular nucleus (PVN) tissues were excised for molecular analyses. FINDINGS Compared with the control, both intravenous and local TMAO administration significantly increased LSG function and activity, shortened effective refractory period, and aggravated ischemia-induced VA. Proinflammatory markers and c-fos in the LSG were also significantly upregulated in both TMAO-treated groups. Particularly, c-fos expression in PVN was significantly increased in the systemic TMAO administration group but not the local TMAO administration group. INTERPRETATION The gut microbe-derived metabolite TMAO can activate the CSNS and aggravate ischemia-induced VA via the direct pathway through the LSG and the indirect pathway through central autonomic activation. FUND: This work was supported by the National Key R&D Program of China [2017YFC1307800], and the National Natural Science Foundation of China [81530011, 81770364, 81570463, 81871486, 81600395, 81600367 and 81700444].
Collapse
Affiliation(s)
- Guannan Meng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Xiaoya Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Liping Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Zhenya Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Meng Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Jielin Deng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Yuhong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Zhen Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Yifeng Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Yanqiu Lai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Qianqian Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Xiaomeng Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Lilei Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China.
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China.
| |
Collapse
|
49
|
Gisslen T, Singh G, Georgieff MK. Fetal inflammation is associated with persistent systemic and hippocampal inflammation and dysregulation of hippocampal glutamatergic homeostasis. Pediatr Res 2019; 85:703-710. [PMID: 30745569 PMCID: PMC6435426 DOI: 10.1038/s41390-019-0330-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 12/29/2018] [Accepted: 02/01/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Inflammation is a major cause of preterm birth and often results in a fetal inflammatory response syndrome (FIRS). Preterm infants with FIRS have a higher childhood incidence of neurodevelopmental disability than preterm infants without FIRS. The mechanisms connecting FIRS to neurodevelopmental disability in formerly preterm infants are not fully understood, but the effect on premature gray matter may have an important role. METHODS Fetal rats were exposed to intra-amniotic (i.a.) LPS 2 days prior to birth to model FIRS. On postnatal day 7, expression of inflammatory mediators was measured in the liver, lung, and brain. Activation of microglia and expression of glutamatergic receptor subunits and transporters were measured in the hippocampus and cortex. RESULTS LPS caused persistent systemic inflammatory mediators gene expression. In the brain, there was corresponding activation of microglia in the hippocampus and cortex. Expression of inflammatory mediators persisted in the hippocampus, but not the cortex, and was associated with altered glutamatergic receptor subunits and transporters. CONCLUSION Hippocampal inflammation and dysregulation of glutamate metabolism persisted well into the postnatal period following i.a. LPS. Poor neurodevelopmental outcomes after FIRS in preterm infants may result in part through glutamatergically driven gray matter injury to the neonatal hippocampus.
Collapse
Affiliation(s)
- Tate Gisslen
- Division of Neonatology, Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA.
| | - Garima Singh
- Division of Neonatology, Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Michael K Georgieff
- Division of Neonatology, Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
50
|
Haque M, Koski KG, Scott ME. Maternal Gastrointestinal Nematode Infection Up-regulates Expression of Genes Associated with Long-Term Potentiation in Perinatal Brains of Uninfected Developing Pups. Sci Rep 2019; 9:4165. [PMID: 30862816 PMCID: PMC6414690 DOI: 10.1038/s41598-019-40729-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 02/19/2019] [Indexed: 12/14/2022] Open
Abstract
Establishment of neural networks critical for memory and cognition begins during the perinatal period but studies on the impact of maternal infection are limited. Using a nematode parasite that remains in the maternal intestine, we tested our hypothesis that maternal infection during pregnancy and early lactation would alter perinatal brain gene expression, and that the anti-inflammatory nature of this parasite would promote synaptic plasticity and long-term potentiation. Brain gene expression was largely unaffected two days after birth, but in seven-day old pups, long-term potentiation and four related pathways essential for the development of synaptic plasticity, cognition and memory were up-regulated in pups of infected dams. Interestingly, our data suggest that a lowering of Th1 inflammatory processes may underscore the apparent beneficial impact of maternal intestinal infection on long-term potentiation.
Collapse
Affiliation(s)
- Manjurul Haque
- Institute of Parasitology, McGill University (Macdonald Campus), 21 111 Lakeshore Road, Ste-Anne-de-Bellevue, Québec, H9X 3V9, Canada
| | - Kristine G Koski
- School of Human Nutrition, McGill University (Macdonald Campus), 21 111 Lakeshore Road, Ste-Anne-de-Bellevue, Québec, H9X 3V9, Canada
| | - Marilyn E Scott
- Institute of Parasitology, McGill University (Macdonald Campus), 21 111 Lakeshore Road, Ste-Anne-de-Bellevue, Québec, H9X 3V9, Canada.
| |
Collapse
|