LIVIVO - The Search Portal for Life Sciences

zur deutschen Oberfläche wechseln
Advanced search

Search results

Result 1 - 10 of total 283

Search options

  1. Article ; Online: Accelerated brain aging with opioid misuse and HIV: New insights on the role of glially derived pro-inflammation mediators and neuronal chloride homeostasis.

    Hauser, Kurt F / Ohene-Nyako, Michael / Knapp, Pamela E

    Current opinion in neurobiology

    2022  Volume 78, Page(s) 102653

    Abstract: Opioid use disorder (OUD) has become a national crisis and contributes to the spread of human immunodeficiency virus (HIV) infection. Emerging evidence and advances in experimental models, methodology, and our understanding of disease processes at the ... ...

    Abstract Opioid use disorder (OUD) has become a national crisis and contributes to the spread of human immunodeficiency virus (HIV) infection. Emerging evidence and advances in experimental models, methodology, and our understanding of disease processes at the molecular and cellular levels reveal that opioids per se can directly exacerbate the pathophysiology of neuroHIV. Despite substantial inroads, the impact of OUD on the severity, development, and prognosis of neuroHIV and HIV-associated neurocognitive disorders is not fully understood. In this review, we explore current evidence that OUD and neuroHIV interact to accelerate cognitive deficits and enhance the neurodegenerative changes typically seen with aging, through their effects on neuroinflammation. We suggest new thoughts on the processes that may underlie accelerated brain aging, including dysregulation of neuronal inhibition, and highlight findings suggesting that opioids, through actions at the μ-opioid receptor, interact with HIV in the central nervous system to promote unique structural and functional comorbid deficits not seen in either OUD or neuroHIV alone.
    MeSH term(s) Humans ; Chlorides ; Inflammation Mediators ; Opioid-Related Disorders ; HIV Infections/complications ; Brain ; Analgesics, Opioid/pharmacology
    Chemical Substances Chlorides ; Inflammation Mediators ; Analgesics, Opioid
    Language English
    Publishing date 2022-12-28
    Publishing country England
    Document type Journal Article ; Review ; Research Support, N.I.H., Extramural
    ZDB-ID 1078046-4
    ISSN 1873-6882 ; 0959-4388
    ISSN (online) 1873-6882
    ISSN 0959-4388
    DOI 10.1016/j.conb.2022.102653
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  2. Article ; Online: HIV-1 Tat and morphine interactions dynamically shift striatal monoamine levels and exploratory behaviors over time.

    Lark, Arianna R S / Nass, Sara R / Hahn, Yun K / Gao, Benlian / Milne, Ginger L / Knapp, Pamela E / Hauser, Kurt F

    Journal of neurochemistry

    2024  Volume 168, Issue 3, Page(s) 185–204

    Abstract: Despite the advent of combination anti-retroviral therapy (cART), nearly half of people infected with HIV treated with cART still exhibit HIV-associated neurocognitive disorders (HAND). HAND can be worsened by co-morbid opioid use disorder. The basal ... ...

    Abstract Despite the advent of combination anti-retroviral therapy (cART), nearly half of people infected with HIV treated with cART still exhibit HIV-associated neurocognitive disorders (HAND). HAND can be worsened by co-morbid opioid use disorder. The basal ganglia are particularly vulnerable to HIV-1 and exhibit higher viral loads and more severe pathology, which can be exacerbated by co-exposure to opioids. Evidence suggests that dopaminergic neurotransmission is disrupted by HIV exposure, however, little is known about whether co-exposure to opioids may alter neurotransmitter levels in the striatum and if this in turn influences behavior. Therefore, we assayed motor, anxiety-like, novelty-seeking, exploratory, and social behaviors, and levels of monoamines and their metabolites following 2 weeks and 2 months of Tat and/or morphine exposure in transgenic mice. Morphine decreased dopamine levels, but significantly elevated norepinephrine, the dopamine metabolites dihydroxyphenylacetic acid (DOPAC) and homovanillic acid (HVA), and the serotonin metabolite 5-hydroxyindoleacetic acid, which typically correlated with increased locomotor behavior. The combination of Tat and morphine altered dopamine, DOPAC, and HVA concentrations differently depending on the neurotransmitter/metabolite and duration of exposure but did not affect the numbers of tyrosine hydroxylase-positive neurons in the mesencephalon. Tat exposure increased the latency to interact with novel conspecifics, but not other novel objects, suggesting the viral protein inhibits exploratory behavior initiation in a context-dependent manner. By contrast, and consistent with prior findings that opioid misuse can increase novelty-seeking behavior, morphine exposure increased the time spent exploring a novel environment. Finally, Tat and morphine interacted to affect locomotor activity in a time-dependent manner, while grip strength and rotarod performance were unaffected. Together, our results provide novel insight into the unique effects of HIV-1 Tat and morphine on monoamine neurochemistry that may underlie their divergent effects on motor and exploratory behavior.
    MeSH term(s) Humans ; Mice ; Animals ; Morphine/pharmacology ; Exploratory Behavior ; HIV-1/metabolism ; Dopamine/metabolism ; 3,4-Dihydroxyphenylacetic Acid/metabolism ; Mice, Transgenic ; Analgesics, Opioid/pharmacology ; Homovanillic Acid ; HIV Infections ; Neurotransmitter Agents ; tat Gene Products, Human Immunodeficiency Virus/metabolism
    Chemical Substances Morphine (76I7G6D29C) ; Dopamine (VTD58H1Z2X) ; 3,4-Dihydroxyphenylacetic Acid (102-32-9) ; Analgesics, Opioid ; Homovanillic Acid (X77S6GMS36) ; Neurotransmitter Agents ; tat Gene Products, Human Immunodeficiency Virus
    Language English
    Publishing date 2024-02-03
    Publishing country England
    Document type Journal Article ; Research Support, N.I.H., Extramural
    ZDB-ID 80158-6
    ISSN 1471-4159 ; 0022-3042 ; 1474-1644
    ISSN (online) 1471-4159
    ISSN 0022-3042 ; 1474-1644
    DOI 10.1111/jnc.16057
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  3. Article ; Online: Fentanyl dysregulates neuroinflammation and disrupts blood-brain barrier integrity in HIV-1 Tat transgenic mice.

    Rademeyer, Kara M / R Nass, Sara / Jones, Austin M / Ohene-Nyako, Michael / Hauser, Kurt F / McRae, MaryPeace

    Journal of neurovirology

    2024  Volume 30, Issue 1, Page(s) 1–21

    Abstract: Opioid overdose deaths have dramatically increased by 781% from 1999 to 2021. In the setting of HIV, opioid drug abuse exacerbates neurotoxic effects of HIV in the brain, as opioids enhance viral replication, promote neuronal dysfunction and injury, and ... ...

    Abstract Opioid overdose deaths have dramatically increased by 781% from 1999 to 2021. In the setting of HIV, opioid drug abuse exacerbates neurotoxic effects of HIV in the brain, as opioids enhance viral replication, promote neuronal dysfunction and injury, and dysregulate an already compromised inflammatory response. Despite the rise in fentanyl abuse and the close association between opioid abuse and HIV infection, the interactive comorbidity between fentanyl abuse and HIV has yet to be examined in vivo. The HIV-1 Tat-transgenic mouse model was used to understand the interactive effects between fentanyl and HIV. Tat is an essential protein produced during HIV that drives the transcription of new virions and exerts neurotoxic effects within the brain. The Tat-transgenic mouse model uses a glial fibrillary acidic protein (GFAP)-driven tetracycline promoter which limits Tat production to the brain and this model is well used for examining mechanisms related to neuroHIV. After 7 days of fentanyl exposure, brains were harvested. Tight junction proteins, the vascular cell adhesion molecule, and platelet-derived growth factor receptor-β were measured to examine the integrity of the blood brain barrier. The immune response was assessed using a mouse-specific multiplex chemokine assay. For the first time in vivo, we demonstrate that fentanyl by itself can severely disrupt the blood-brain barrier and dysregulate the immune response. In addition, we reveal associations between inflammatory markers and tight junction proteins at the blood-brain barrier.
    MeSH term(s) Animals ; Blood-Brain Barrier/drug effects ; Blood-Brain Barrier/metabolism ; Blood-Brain Barrier/pathology ; Blood-Brain Barrier/virology ; Mice, Transgenic ; Mice ; Fentanyl/pharmacology ; HIV-1/drug effects ; HIV-1/genetics ; tat Gene Products, Human Immunodeficiency Virus/genetics ; tat Gene Products, Human Immunodeficiency Virus/metabolism ; Neuroinflammatory Diseases/genetics ; Neuroinflammatory Diseases/pathology ; Neuroinflammatory Diseases/virology ; HIV Infections/virology ; HIV Infections/genetics ; HIV Infections/pathology ; HIV Infections/drug therapy ; Disease Models, Animal ; Analgesics, Opioid/pharmacology ; Analgesics, Opioid/adverse effects ; Glial Fibrillary Acidic Protein/genetics ; Glial Fibrillary Acidic Protein/metabolism ; Tight Junction Proteins/metabolism ; Tight Junction Proteins/genetics ; Humans ; Brain/drug effects ; Brain/virology ; Brain/metabolism ; Brain/pathology ; Opioid-Related Disorders/genetics ; Opioid-Related Disorders/pathology ; Opioid-Related Disorders/metabolism
    Chemical Substances Fentanyl (UF599785JZ) ; tat Gene Products, Human Immunodeficiency Virus ; Analgesics, Opioid ; Glial Fibrillary Acidic Protein ; Tight Junction Proteins
    Language English
    Publishing date 2024-01-27
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural
    ZDB-ID 1283265-0
    ISSN 1538-2443 ; 1355-0284
    ISSN (online) 1538-2443
    ISSN 1355-0284
    DOI 10.1007/s13365-023-01186-4
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  4. Article: Opiate Drugs with Abuse Liability Hijack the Endogenous Opioid System to Disrupt Neuronal and Glial Maturation in the Central Nervous System.

    Hauser, Kurt F / Knapp, Pamela E

    Frontiers in pediatrics

    2018  Volume 5, Page(s) 294

    Abstract: The endogenous opioid system, comprised of multiple opioid neuropeptide and receptor gene families, is highly expressed by developing neural cells and can significantly influence neuronal and glial maturation. In many central nervous system (CNS) regions, ...

    Abstract The endogenous opioid system, comprised of multiple opioid neuropeptide and receptor gene families, is highly expressed by developing neural cells and can significantly influence neuronal and glial maturation. In many central nervous system (CNS) regions, the expression of opioid peptides and receptors occurs only transiently during development, effectively disappearing with subsequent maturation only to reemerge under pathologic conditions, such as with inflammation or injury. Opiate drugs with abuse liability act to modify growth and development by mimicking the actions of endogenous opioids. Although typically mediated by μ-opioid receptors, opiate drugs can also act through δ- and κ-opioid receptors to modulate growth in a cell-type, region-specific, and developmentally regulated manner. Opioids act as biological response modifiers and their actions are highly contextual, plastic, modifiable, and influenced by other physiological processes or pathophysiological conditions, such as neuro-acquired immunodeficiency syndrome. To date, most studies have considered the acute effects of opiates on cellular maturation. For example, activating opioid receptors typically results in acute growth inhibition in both neurons and glia. However, with sustained opioid exposure, compensatory factors become operative, a concept that has been largely overlooked during CNS maturation. Accordingly, this article surveys prior studies on the effects of opiates on CNS maturation, and also suggests new directions for future research in this area. Identifying the cellular and molecular mechanisms underlying the adaptive responses to chronic opiate exposure (e.g., tolerance) during maturation is crucial toward understanding the consequences of perinatal opiate exposure on the CNS.
    Language English
    Publishing date 2018-01-23
    Publishing country Switzerland
    Document type Journal Article ; Review
    ZDB-ID 2711999-3
    ISSN 2296-2360
    ISSN 2296-2360
    DOI 10.3389/fped.2017.00294
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  5. Article ; Online: Opioid and neuroHIV Comorbidity - Current and Future Perspectives.

    Fitting, Sylvia / McRae, MaryPeace / Hauser, Kurt F

    Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology

    2020  Volume 15, Issue 4, Page(s) 584–627

    Abstract: With the current national opioid crisis, it is critical to examine the mechanisms underlying pathophysiologic interactions between human immunodeficiency virus (HIV) and opioids in the central nervous system (CNS). Recent advances in experimental models, ...

    Abstract With the current national opioid crisis, it is critical to examine the mechanisms underlying pathophysiologic interactions between human immunodeficiency virus (HIV) and opioids in the central nervous system (CNS). Recent advances in experimental models, methodology, and our understanding of disease processes at the molecular and cellular levels reveal opioid-HIV interactions with increasing clarity. However, despite the substantial new insight, the unique impact of opioids on the severity, progression, and prognosis of neuroHIV and HIV-associated neurocognitive disorders (HAND) are not fully understood. In this review, we explore, in detail, what is currently known about mechanisms underlying opioid interactions with HIV, with emphasis on individual HIV-1-expressed gene products at the molecular, cellular and systems levels. Furthermore, we review preclinical and clinical studies with a focus on key considerations when addressing questions of whether opioid-HIV interactive pathogenesis results in unique structural or functional deficits not seen with either disease alone. These considerations include, understanding the combined consequences of HIV-1 genetic variants, host variants, and μ-opioid receptor (MOR) and HIV chemokine co-receptor interactions on the comorbidity. Lastly, we present topics that need to be considered in the future to better understand the unique contributions of opioids to the pathophysiology of neuroHIV. Graphical Abstract Blood-brain barrier and the neurovascular unit. With HIV and opiate co-exposure (represented below the dotted line), there is breakdown of tight junction proteins and increased leakage of paracellular compounds into the brain. Despite this, opiate exposure selectively increases the expression of some efflux transporters, thereby restricting brain penetration of specific drugs.
    MeSH term(s) AIDS Dementia Complex/complications ; COVID-19 ; HIV Infections/complications ; HIV-1/immunology ; Humans ; Opioid Epidemic ; Opioid-Related Disorders/epidemiology
    Keywords covid19
    Language English
    Publishing date 2020-09-02
    Publishing country United States
    Document type Journal Article ; Review
    ZDB-ID 2227405-4
    ISSN 1557-1904 ; 1557-1890
    ISSN (online) 1557-1904
    ISSN 1557-1890
    DOI 10.1007/s11481-020-09941-8
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  6. Article: Neurodegeneration Within the Amygdala Is Differentially Induced by Opioid and HIV-1 Tat Exposure.

    Nass, Sara R / Ohene-Nyako, Michael / Hahn, Yun K / Knapp, Pamela E / Hauser, Kurt F

    Frontiers in neuroscience

    2022  Volume 16, Page(s) 804774

    Abstract: Opioid use disorder (OUD) is a critical problem that contributes to the spread of HIV and may intrinsically worsen neuroHIV. Despite the advent of combined antiretroviral therapies (cART), about half of persons infected with HIV (PWH) experience ... ...

    Abstract Opioid use disorder (OUD) is a critical problem that contributes to the spread of HIV and may intrinsically worsen neuroHIV. Despite the advent of combined antiretroviral therapies (cART), about half of persons infected with HIV (PWH) experience cognitive and emotional deficits that can be exacerbated by opioid abuse. HIV-1 Tat is expressed in the central nervous system (CNS) of PWH on cART and is thought to contribute to neuroHIV. The amygdala regulates emotion and memories associated with fear and stress and is important in addiction behavior. Notwithstanding its importance in emotional saliency, the effects of HIV and opioids in the amygdala are underexplored. To assess Tat- and morphine-induced neuropathology within the amygdala, male Tat transgenic mice were exposed to Tat for 8 weeks and administered saline and/or escalating doses of morphine twice daily (s.c.) during the last 2 weeks of Tat exposure. Eight weeks of Tat exposure decreased the acoustic startle response and the dendritic spine density in the basolateral amygdala, but not the central nucleus of the amygdala. In contrast, repeated exposure to morphine alone, but not Tat, increased the acoustic startle response and whole amygdalar levels of amyloid-β (Aβ) monomers and oligomers and tau phosphorylation at Ser396, but not neurofilament light chain levels. Co-exposure to Tat and morphine decreased habituation and prepulse inhibition to the acoustic startle response and potentiated the morphine-induced increase in Aβ monomers. Together, our findings indicate that sustained Tat and morphine exposure differentially promote synaptodendritic degeneration within the amygdala and alter sensorimotor processing.
    Language English
    Publishing date 2022-05-04
    Publishing country Switzerland
    Document type Journal Article
    ZDB-ID 2411902-7
    ISSN 1662-453X ; 1662-4548
    ISSN (online) 1662-453X
    ISSN 1662-4548
    DOI 10.3389/fnins.2022.804774
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  7. Article ; Online: HIV-1 Tat reduces apical dendritic spine density throughout the trisynaptic pathway in the hippocampus of male transgenic mice.

    McLane, Virginia D / Lark, Arianna R S / Nass, Sara R / Knapp, Pamela E / Hauser, Kurt F

    Neuroscience letters

    2022  Volume 782, Page(s) 136688

    Abstract: Nearly one-third of persons infected with HIV-1 (PWH) develop HIV-associated neurocognitive disorders (HAND), which can be exacerbated by exposure to opioids. The impact of opioids on HIV-induced alterations in neuronal plasticity is less well understood. ...

    Abstract Nearly one-third of persons infected with HIV-1 (PWH) develop HIV-associated neurocognitive disorders (HAND), which can be exacerbated by exposure to opioids. The impact of opioids on HIV-induced alterations in neuronal plasticity is less well understood. Both morphine exposure and HIV have been shown to disrupt synaptic growth and stability in the hippocampus suggesting a potential site of convergence for their deleterious effects. In the present study, we examined the density of dendritic spines in CA1 and CA3 pyramidal neurons, and granule neurons within the dentate gyrus representing the hippocampal trisynaptic pathway after short-term exposure to the HIV transactivator of transcription (Tat) protein and morphine. We exposed inducible male, HIV-1 Tat transgenic mice to escalating doses of morphine (10-40 mg/kg, b.i.d.) and examined synaptodendritic structure in Golgi-impregnated hippocampal neurons. HIV-1 Tat, but not morphine, systematically reduced the density of apical, but not basilar, dendrites of CA1 and CA3 pyramidal neurons, and granule neuronal apical dendrites, suggesting the coordinated loss of specific synaptic interconnections throughout the hippocampal trisynaptic pathway.
    MeSH term(s) Analgesics, Opioid/pharmacology ; Animals ; Dendrites/metabolism ; Dendritic Spines ; HIV-1 ; Hippocampus ; Male ; Mice ; Mice, Transgenic ; Morphine/metabolism ; Morphine/pharmacology
    Chemical Substances Analgesics, Opioid ; Morphine (76I7G6D29C)
    Language English
    Publishing date 2022-05-17
    Publishing country Ireland
    Document type Journal Article ; Research Support, N.I.H., Extramural
    ZDB-ID 194929-9
    ISSN 1872-7972 ; 0304-3940
    ISSN (online) 1872-7972
    ISSN 0304-3940
    DOI 10.1016/j.neulet.2022.136688
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  8. Article ; Online: Myelin regulatory factor is a target of individual and interactive effects of HIV-1 Tat and morphine in the striatum and pre-frontal cortex.

    Flounlacker, Kelly M / Hahn, Yun Kyung / Xu, Ruqiang / Simons, Chloe A / Tian, Tao / Hauser, Kurt F / Knapp, Pamela E

    Journal of neurovirology

    2023  Volume 29, Issue 1, Page(s) 15–26

    Abstract: HIV-associated neurocognitive disorders (HAND) remain pervasive even with increased efficacy/use of antiretroviral therapies. Opioid use/abuse among HIV + individuals is documented to exacerbate CNS deficits. White matter (WM) alterations, including ... ...

    Abstract HIV-associated neurocognitive disorders (HAND) remain pervasive even with increased efficacy/use of antiretroviral therapies. Opioid use/abuse among HIV + individuals is documented to exacerbate CNS deficits. White matter (WM) alterations, including myelin pallor, and volume/structural alterations detected by diffusion tensor imaging are common observations in HIV + individuals, and studies in non-human primates suggest that WM may harbor virus. Using transgenic mice that express the HIV-1 Tat protein, we examined in vivo effects of 2-6 weeks of Tat and morphine exposure on WM using genomic and biochemical methods. RNA sequencing of striatal tissue at 2 weeks revealed robust changes in mRNAs associated with oligodendrocyte precursor populations and myelin integrity, including those for transferrin, the atypical oligodendrocyte marker N-myc downstream regulated 1 (Ndrg1), and myelin regulatory factor (Myrf/Mrf), an oligodendrocyte-specific transcription factor with a significant role in oligodendrocyte differentiation/maturation. Western blots conducted after 6-weeks exposure in 3 brain regions (striatum, corpus callosum, pre-frontal cortex) revealed regional differences in the effect of Tat and morphine on Myrf levels, and on levels of myelin basic protein (MBP), whose transcription is regulated by Myrf. Responses included individual and interactive effects. Although baseline and post-treatment levels of Myrf and MBP differed between brain regions, post-treatment MBP levels in striatum and pre-frontal cortex were compatible with changes in Myrf activity. Additionally, the Myrf regulatory ubiquitin ligase Fbxw7 was identified as a novel target in our model. These results suggest that Myrf and Fbxw7 contribute to altered myelin gene regulation in HIV.
    MeSH term(s) Animals ; Mice ; Diffusion Tensor Imaging ; F-Box-WD Repeat-Containing Protein 7/metabolism ; Frontal Lobe/metabolism ; HIV Infections ; HIV-1/metabolism ; Mice, Transgenic ; Morphine ; Transcription Factors/genetics ; tat Gene Products, Human Immunodeficiency Virus/metabolism
    Chemical Substances F-Box-WD Repeat-Containing Protein 7 ; Morphine (76I7G6D29C) ; Transcription Factors ; tat Gene Products, Human Immunodeficiency Virus
    Language English
    Publishing date 2023-02-28
    Publishing country United States
    Document type Journal Article
    ZDB-ID 1283265-0
    ISSN 1538-2443 ; 1355-0284
    ISSN (online) 1538-2443
    ISSN 1355-0284
    DOI 10.1007/s13365-022-01107-x
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  9. Article ; Online: Restoration of KCC2 Membrane Localization in Striatal Dopamine D2 Receptor-Expressing Medium Spiny Neurons Rescues Locomotor Deficits in HIV Tat-Transgenic Mice.

    Barbour, Aaron J / Nass, Sara R / Hahn, Yun K / Hauser, Kurt F / Knapp, Pamela E

    ASN neuro

    2021  Volume 13, Page(s) 17590914211022089

    Abstract: People infected with HIV (PWH) are highly susceptible to striatal and hippocampal damage. Motor and memory impairments are common among these patients, likely as behavioral manifestations of damage to these brain regions. GABAergic dysfunction from HIV ... ...

    Abstract People infected with HIV (PWH) are highly susceptible to striatal and hippocampal damage. Motor and memory impairments are common among these patients, likely as behavioral manifestations of damage to these brain regions. GABAergic dysfunction from HIV infection and viral proteins such as transactivator of transcription (Tat) have been well documented. We recently demonstrated that the neuron specific Cl
    MeSH term(s) Animals ; Corpus Striatum/metabolism ; HIV Infections ; Mice ; Mice, Inbred C57BL ; Mice, Transgenic ; Neurons/metabolism ; Receptors, Dopamine D1/genetics ; Receptors, Dopamine D1/metabolism ; Receptors, Dopamine D2/genetics ; Receptors, Dopamine D2/metabolism ; Symporters/genetics ; Symporters/metabolism ; Trans-Activators ; tat Gene Products, Human Immunodeficiency Virus ; K Cl- Cotransporters
    Chemical Substances DRD2 protein, mouse ; Receptors, Dopamine D1 ; Receptors, Dopamine D2 ; Symporters ; Trans-Activators ; tat Gene Products, Human Immunodeficiency Virus
    Language English
    Publishing date 2021-08-18
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural
    ZDB-ID 2485467-0
    ISSN 1759-0914 ; 1759-0914
    ISSN (online) 1759-0914
    ISSN 1759-0914
    DOI 10.1177/17590914211022089
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  10. Article ; Online: Casein Kinase 2 Mediates HIV- and Opioid-Induced Pathologic Phosphorylation of TAR DNA Binding Protein 43 in the Basal Ganglia.

    Ohene-Nyako, Michael / Nass, Sara R / Richard, Hope T / Lukande, Robert / Nicol, Melanie R / McRae, MaryPeace / Knapp, Pamela E / Hauser, Kurt F

    ASN neuro

    2023  Volume 15, Page(s) 17590914231158218

    Abstract: Summary statement: HIV/HIV-1 Tat and morphine independently increase pathologic phosphorylation of TAR DNA binding protein 43 in the striatum. HIV- and opioid-induced pathologic phosphorylation of TAR DNA binding protein 43 may involve enhanced CK2 ... ...

    Abstract Summary statement: HIV/HIV-1 Tat and morphine independently increase pathologic phosphorylation of TAR DNA binding protein 43 in the striatum. HIV- and opioid-induced pathologic phosphorylation of TAR DNA binding protein 43 may involve enhanced CK2 activity and protein levels.
    MeSH term(s) Humans ; Phosphorylation ; Casein Kinase II/metabolism ; Analgesics, Opioid/pharmacology ; Analgesics, Opioid/metabolism ; tat Gene Products, Human Immunodeficiency Virus/metabolism ; DNA-Binding Proteins ; HIV-1/metabolism ; Basal Ganglia/metabolism ; HIV Infections ; Protein Binding
    Chemical Substances Casein Kinase II (EC 2.7.11.1) ; Analgesics, Opioid ; tat Gene Products, Human Immunodeficiency Virus ; DNA-Binding Proteins
    Language English
    Publishing date 2023-03-08
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural
    ZDB-ID 2485467-0
    ISSN 1759-0914 ; 1759-0914
    ISSN (online) 1759-0914
    ISSN 1759-0914
    DOI 10.1177/17590914231158218
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

To top