Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 29
1.
Eur J Neurosci ; 56(2): 3806-3824, 2022 07.
Article En | MEDLINE | ID: mdl-35543645

It is widely accepted that the surface glycoprotein (gp120) of human immunodeficiency virus-1 (HIV-1) plays an important role in HIV-1-induced nerve damage and pathogenesis of HIV-associated neurocognitive disorders (HAND). Our previous work has demonstrated that gp120 enhanced excitatory postsynaptic currents (EPSCs) mediated by N-methyl-d-aspartate receptors (NMDARs) and caused neural injury. However, the relationship between gp120, NMDARs and HAND is still unclear. Several lines of evidence indicate that double-stranded RNA-activated protein kinase (PKR) is involved in NMDA-induced cerebral ischaemia and retinal damage, but because its role in neuropathology is still debated, we examined whether PKR links oxidative stress and endoplasmic reticulum (ER) stress to exert a deleterious role in the rat model with gp120-induced dementia. In this study, we found that NMDAR antagonist memantine or PKR inhibitor C16 improved gp120-induced learning and memory impairment and inhibited gp120-induced PKR activity. Furthermore, memantine or C16 was found to attenuate gp120-induced neuroinflammation, oxidative stress, ER stress and its downstream IRE1α/JNK pathway. Additionally, memantine or C16 evidently inhibited apoptotic pathways by reducing the Bax and caspase-3, -8, -9 expressions and increasing Bcl-2 expression. So the NMDA receptor antagonists could alleviate HIV/gp120-induced dementia in the rat model by altering PKR level. In conclusion, this study demonstrates that NMDARs play a key role in HIV/gp120-induced hippocampal damage and cognitive dysfunction through PKR-mediated oxidative stress, ER stress, and IRE1α/JNK signalling pathway in rats, and implicating PKR inhibitors could provide a novel neuroprotective strategy for HAND via inhibiting ER stress and its downstream IRE1α signalling pathway.


Cognitive Dysfunction , Dementia , HIV Envelope Protein gp120 , Neuroprotection , Receptors, N-Methyl-D-Aspartate , Animals , Apoptosis , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Endoplasmic Reticulum Stress , Endoribonucleases/metabolism , Endoribonucleases/pharmacology , HIV Envelope Protein gp120/adverse effects , Humans , Memantine/pharmacology , Oxidative Stress , Protein Serine-Threonine Kinases , Rats , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Signal Transduction
2.
Mol Pain ; 16: 1744806920922100, 2020.
Article En | MEDLINE | ID: mdl-32354292

HIV-associated neuropathic pain (HNP) is a common complication for AIDS patients. The pathological mechanism governing HNP has not been elucidated, and HNP has no effective analgesic treatment. Brain-derived neurotrophic factor (BDNF) is a member of the neurotrophic factor family related to the plasticity of the central nervous system. BDNF dysregulation is involved in many neurological diseases, including neuropathic pain. However, to the best of our knowledge, the role and mechanism of BDNF in HNP have not been elucidated. In this study, we explored this condition in an HNP mouse model induced by intrathecal injection of gp120. We found that Wnt3a and ß-catenin expression levels increased in the spinal cord of HNP mice, consequently regulating the expression of BDNF and affecting hypersensitivity. In addition, the blockade of Wing-Int/ß-catenin signaling, BDNF/TrkB or the BDNF/p75NTR pathway alleviated mechanical allodynia. BDNF immunoreactivity was colocalized with spinal microglial cells, which were activated in HNP mice. Inhibition of spinal microglial cell activation by minocycline relieved mechanical allodynia in HNP mice. This study helped to elucidate the role of the Wing-Int/ß-catenin/BDNF signaling axis in HNP and may establish a foundation for further research investigating the Wing-Int/ß-catenin/BDNF signaling axis as a target for HNP treatment.


Brain-Derived Neurotrophic Factor/metabolism , HIV Envelope Protein gp120/adverse effects , Microglia/metabolism , Neuralgia/metabolism , Spinal Cord/pathology , Wnt Signaling Pathway , Animals , Behavior, Animal , HIV Envelope Protein gp120/administration & dosage , Hyperalgesia/complications , Hyperalgesia/pathology , Injections, Spinal , Mice, Inbred ICR , Models, Biological , Neuralgia/complications , Nociception , Receptor, trkB/metabolism , Receptors, Nerve Growth Factor/metabolism , Up-Regulation
3.
Brain Behav Immun ; 88: 325-339, 2020 08.
Article En | MEDLINE | ID: mdl-32229220

The neuroinflammatory responses to human immunodeficiency virus type 1 (HIV-1) coat proteins, such as glycoprotein 120 (gp120), are considered to be responsible for the HIV-associated distal sensory neuropathy. Accumulating evidences suggest that T-cell line tropic X4 gp120 increases macrophage infiltration into the peripheral nerves, and thereby induces neuroinflammation leading to pain. However, the mechanisms underlying X4 gp120-induced macrophage recruitment to the peripheral nervous systems remain unclear. Here, we demonstrated that perineural application of X4 gp120 from HIV-1 strains IIIB and MN elicited mechanical hypersensitivity and spontaneous pain-like behaviors in mice. Furthermore, flow cytometry and immunohistochemical studies revealed increased infiltration of bone marrow-derived macrophages into the parenchyma of sciatic nerves and dorsal root ganglia (DRG) 7 days after gp120 IIIB or MN application. Chemical deletion of circulating macrophages using clodronate liposomes markedly suppressed gp120 IIIB-induced pain-like behaviors. In in vitro cell infiltration analysis, RAW 264.7 cell (a murine macrophage cell line) was chemoattracted to conditioned medium from gp120 IIIB- or MN-treated cultured Schwann cells, but not to conditioned medium from these gp120-treated DRG neurons, suggesting possible involvement of Schwann cell-derived soluble factors in macrophage infiltration. We identified using a gene expression array that CXCL1, a chemoattractant of macrophages and neutrophils, was increased in gp120 IIIB-treated cultured Schwann cells. Similar to gp120 IIIB or MN, perineural application of recombinant CXCL1 elicited pain-like behaviors accompanied by macrophage infiltration to the peripheral nerves. Furthermore, the repeated injection of CXCR2 (receptor for CXCL1) antagonist or CXCL1 neutralizing antibody prevented both pain-like behaviors and macrophage infiltration in gp120 IIIB-treated mice. Thus, the present study newly defines that Schwann cell-derived CXCL1, secreted in response to X4 gp120 exposure, is responsible for macrophage infiltration into peripheral nerves, and is thereby associated with pain-like behaviors in mice. We propose herein that communication between Schwann cells and macrophages may play a prominent role in the induction of X4 HIV-1-associated pain.


Chemokine CXCL1/metabolism , HIV Envelope Protein gp120/adverse effects , Macrophages/cytology , Neuralgia , Schwann Cells/metabolism , Animals , Glycoproteins , HIV-1 , Mice
4.
Lancet HIV ; 6(11): e737-e749, 2019 11.
Article En | MEDLINE | ID: mdl-31601541

BACKGROUND: Up to now, immunisation regimens that have been assessed for development of HIV vaccines have included purified envelope (Env) protein among the boosting components of the regimen. We postulated that co-administration of Env protein with either a DNA or NYVAC vector during priming would result in early generation of antibody responses to the Env V1/V2 region, which are important markers for effective protection against infection. We aimed to assess the safety and immunogenicity of a multivalent HIV vaccine including either DNA or NYVAC vectors alone or in combination with Env glycoprotein (gp120) followed by a co-delivered NYVAC and Env protein boost. METHODS: We did a single-centre, double-blind, placebo-controlled phase 1b trial at the Centre Hospitalier Universitaire Vaudois (Lausanne, Switzerland). We included healthy volunteers aged 18-50 years who were at low risk of HIV infection. We randomly allocated participants using computer-generated random numbers to one of four vaccination schedules or placebo (4:1), and within these schedules participants were allocated either active treatment (T1, T2, T3, and T4) or placebo (C1, C2, C3, and C4). T1 consisted of two doses of NYVAC vector followed by two doses of NYVAC vector and gp120 Env protein; T2 comprised four doses of NYVAC vector and gp120 Env protein; T3 was two doses of DNA vector followed by two doses of NYVAC vector and gp120 Env protein; and T4 was two doses of DNA vector and gp120 Env protein followed by two doses of NYVAC vector and gp120 Env protein. Placebo injections were matched to the corresponding active treatment group. Doses were administered by injection at months 0, 1, 3, and 6. Primary outcomes were safety and immunogenicity of the vaccine schedules. Immune response measures included cross-clade and epitope-specific binding antibodies, neutralising antibodies, and antibody-dependent cell-mediated cytotoxicity measured 2 weeks after the month 1, 3, and 6 vaccinations. This trial is registered with ClinicalTrials.gov, NCT01799954. FINDINGS: Between Aug 23, 2012, and April 18, 2013, 148 healthy adult volunteers were screened for the trial, of whom 96 participants were enrolled. 20 individuals were allocated to each active treatment group (groups T1-4; n=80) and four were assigned to each placebo group (groups C1-4; n=16). Vaccines containing the NYVAC vector (groups T1 and T2) were associated with more frequent severe reactogenicity and more adverse events than were vaccines containing the DNA vector (groups T3 and T4). The most frequent adverse events judged related to study product were lymphadenopathy (n=9) and hypoaesthesia (n=2). Two participants, one in the placebo group and one in the DNA-primed T3 group, had serious adverse events that were judged unrelated to study product. One participant in the T3 group died from cranial trauma after a motor vehicle accident. Across the active treatment groups, IgG responses 2 weeks after the 6-month dose of vaccine were 74-95%. Early administration of gp120 Env protein (groups T2 and T4) was associated with a substantially earlier and higher area under the curve for gp120 Env binding, production of anti-V1/V2 and neutralising antibodies, and better antibody-response coverage over a period of 18 months, compared with vaccination regimens that delayed administration of gp120 Env protein until the 3-month vaccination (groups T1 and T3). INTERPRETATION: Co-administration of gp120 Env protein components with DNA or NYVAC vectors during priming led to early and potent induction of Env V1/V2 IgG binding antibody responses. This immunisation approach should be considered for induction of preventive antibodies in future HIV vaccine efficacy trials. FUNDING: National Institutes of Health, National Institute of Allergy and Infectious Diseases, and the Bill & Melinda Gates Foundation.


AIDS Vaccines/administration & dosage , Antibodies, Neutralizing/metabolism , HIV Antibodies/metabolism , HIV Envelope Protein gp120/administration & dosage , HIV Infections/prevention & control , Vaccines, DNA/administration & dosage , AIDS Vaccines/adverse effects , AIDS Vaccines/immunology , Adult , Area Under Curve , Double-Blind Method , Drug Administration Schedule , Drug Therapy, Combination , Female , HIV Envelope Protein gp120/adverse effects , HIV Envelope Protein gp120/immunology , HIV Infections/immunology , Humans , Male , Middle Aged , Vaccines, DNA/adverse effects , Vaccines, DNA/immunology , Young Adult
6.
J Acquir Immune Defic Syndr ; 63(1): 1-8, 2013 May 01.
Article En | MEDLINE | ID: mdl-23221981

BACKGROUND: The development of a safe and effective vaccine against HIV type 1 for the prevention of mother-to-child transmission of HIV would significantly advance the goal of eliminating HIV infection in children. Safety and feasibility results from phase 1, randomized, double-blind, placebo-controlled trial of ALVAC-HIV vCP1521 in infants born to HIV type 1-infected women in Uganda are reported. METHODS: HIV-exposed infants were enrolled at birth and randomized (4:1) to receive vaccine or saline placebo intramuscular injections at birth, 4, 8, and 12 weeks of age. Vaccine reactogenicity was assessed at vaccination and days 1 and 2 postvaccination. Infants were followed until 24 months of age. HIV infection status was determined by HIV DNA polymerase chain reaction. RESULTS: From October 2006 to May 2007, 60 infants (48 vaccine and 12 placebo) were enrolled with 98% retention at 24 months. One infant was withdrawn, but there were no missed visits or vaccinations among the 59 infants retained. Immune responses elicited by diphtheria, polio, hepatitis B, haemophilus influenzae type B, and measles vaccination were similar in the 2 arms. The vaccine was well tolerated with no severe or life-threatening reactogenicity events. Adverse events were equally distributed across both study arms. Four infants were diagnosed as HIV infected [3 at birth (2 vaccine and 1 placebo) and 1 in vaccine arm at 2 weeks of age]. CONCLUSION: The ALVAC-HIV vCP1521 vaccination was feasible and safe in infants born to HIV-infected women in Uganda. The conduct of high-quality infant HIV vaccine trials is achievable in Africa.


AIDS Vaccines , HIV Envelope Protein gp120 , HIV Infections/prevention & control , Infectious Disease Transmission, Vertical/prevention & control , AIDS Vaccines/administration & dosage , AIDS Vaccines/adverse effects , Adolescent , Adult , Breast Feeding , Child, Preschool , Double-Blind Method , Female , HIV Envelope Protein gp120/administration & dosage , HIV Envelope Protein gp120/adverse effects , HIV Infections/diagnosis , HIV Infections/transmission , HIV Infections/virology , HIV-1 , Humans , Infant , Infant, Newborn , Milk, Human , Treatment Outcome , Uganda , Vaccination , Young Adult
7.
Eur J Pain ; 16(10): 1357-67, 2012 Nov.
Article En | MEDLINE | ID: mdl-22467279

BACKGROUND: Peripheral nerve injury is associated with a spinal microglial response that has been correlated with the development of behaviours reflective of neuropathic pain. METHODS: To examine whether this phenomenon is generalizable to neuropathic pain of non-traumatic aetiology, this study investigated the association between spinal microgliosis and behavioural measures of neuropathic hypersensitivity and pain-related anxiety behaviour in four distinct rat models of peripheral neuropathic pain. These were traumatic neuropathy [L5 spinal nerve transection (SNT)], HIV-related neuropathies (either treatment with the antiretroviral drug Zalcitabine (ddC) or combination of perineural exposure to the HIV-gp120 protein and ddC treatment) and varicella zoster virus (VZV) infection. RESULTS AND CONCLUSION: Persistent mechanical hypersensitivity developed in all 'neuropathic' rats. However, spinal microgliosis, as measured by increased CD11b/c immunohistochemical staining and increased numbers of cells expressing CD11b measured by flow cytometry, was evident in the SNT and to a lesser extent in the HIV neuropathy models but not the VZV model. These results suggest that behavioural hypersensitivity and thigmotaxis can only be linked to a microglial response in certain models of neuropathy.


Behavior, Animal , Gliosis/pathology , Microglia/pathology , Peripheral Nervous System Diseases/pathology , Spinal Cord/pathology , Animals , Anti-HIV Agents/adverse effects , Disease Models, Animal , Flow Cytometry , HIV Envelope Protein gp120/adverse effects , HIV Infections/complications , HIV Infections/pathology , Herpes Zoster/complications , Herpes Zoster/pathology , Herpesvirus 3, Human , Hyperalgesia/pathology , Immunohistochemistry , Male , Peripheral Nerve Injuries/complications , Peripheral Nerve Injuries/pathology , Peripheral Nervous System Diseases/etiology , Rats , Rats, Wistar , Spinal Nerves/injuries , Zalcitabine/adverse effects
8.
PLoS One ; 6(12): e27837, 2011.
Article En | MEDLINE | ID: mdl-22205930

BACKGROUND: A prime-boost vaccination regimen with ALVAC-HIV (vCP1521) administered intramuscularly at 0, 4, 12, and 24 weeks and gp120 AIDSVAX B/E at 12 and 24 weeks demonstrated modest efficacy of 31.2% for prevention of HIV acquisition in HIV-uninfected adults participating in a community-based efficacy trial in Thailand. METHODOLOGY/PRINCIPAL FINDINGS: Reactogenicity was recorded for 3 days following vaccination. Adverse events were monitored every 6 months for 3.5 years, during which pregnancy outcomes were recorded. Of the 16,402 volunteers, 69% of the participants reported an adverse event any time after the first dose. Only 32.9% experienced an AE within 30 days following any vaccination. Overall adverse event rates and attribution of relatedness did not differ between groups. The frequency of serious adverse events was similar in vaccine (14.3%) and placebo (14.9%) recipients (p = 0.33). None of the 160 deaths (85 in vaccine and 75 in placebo recipients, p = 0.43) was assessed as related to vaccine. The most common cause of death was trauma or traffic accident. Approximately 30% of female participants reported a pregnancy during the study. Abnormal pregnancy outcomes were experienced in 17.1% of vaccine and 14.6% (p = 0.13) of placebo recipients. When the conception occurred within 3 months (estimated) of a vaccination, the majority of these abnormal outcomes were spontaneous or elective abortions among 22.2% and 15.3% of vaccine and placebo pregnant recipients, respectively (p = 0.08). Local reactions occurred in 88.0% of vaccine and 61.0% of placebo recipients (p<0.001) and were more frequent after ALVAC-HIV than AIDSVAX B/E vaccination. Systemic reactions were more frequent in vaccine than placebo recipients (77.2% vs. 59.8%, p<0.001). Local and systemic reactions were mostly mild to moderate, resolving within 3 days. CONCLUSIONS/SIGNIFICANCE: The ALVAC-HIV and AIDSVAX B/E vaccine regimen was found to be safe, well tolerated and suitable for potential large-scale use in Thailand. TRIAL REGISTRATION: ClinicalTrials.govNCT00223080.


AIDS Vaccines/adverse effects , AIDS Vaccines/immunology , HIV Envelope Protein gp120/immunology , HIV-1/immunology , Immunization, Secondary/adverse effects , Safety , Vaccination/adverse effects , Adolescent , Adult , Female , HIV Envelope Protein gp120/adverse effects , Humans , Male , Pregnancy , Pregnancy Outcome , Thailand , Young Adult
9.
Rev. neurol. (Ed. impr.) ; 52(2): 101-111, 16 ene., 2011. tab, ilus
Article Es | IBECS | ID: ibc-86969

La neuroinflamación constituye un proceso clave en la neuropatogénesis del virus del sida como consecuencia de la activación aberrante de receptores de quimiocinas (CXCR4, CX3CR1 y CCR5), ya que la liberación de citocinas proinflamatorias por las células infectadas amplifica la neurotoxicidad microglial y genera lipoperóxidos y especies reactivas de oxígeno que, en última instancia, dañan la neurona. Por otro lado, la neurotoxina Tat induce alteraciones dendríticas por interacción con el receptor LRP (receptor de lipoproteínas de baja densidad) e induce una excesiva estimulación de los receptores de N-metil D-aspartato. Además, la interacción aberrante de la glucoproteína gp120 con el receptor CXCR4 induce apoptosis dependiente de caspasa 3 (también libera ceramida) y activa las proteínas apoptóticas p53 y retinoblastoma como mecanismos neurotóxicos asociados a la disfunción neural en el virus de la inmunodeficiencia humana 1 (VIH-1). Asimismo, la gliosis/activación microglial y la liberación de factores virales por los monocitos infectados, y el incremento de determinadas quimiocinas en el líquido cefalorraquídeo (MCP-1 y fractalcina, entre otras), contribuyen a la neuropatogénesis del VIH-1. Por otro lado, se han detectado depósitos de alfa-sinucleína y de beta-amiloide en cerebros post mortem de seropositivos de edad avanzada. Además, se han descrito varios marcadores sistémicos relacionados con los efectos degenerativos del virus y de sus neurotoxinas en el sistema nervioso central, tales como osteopontina, CD163 y fractalcina, entre otros. Por último, se han realizado ensayos clínicos basados en estrategias protectoras relacionadas con la inhibición de proteínas apoptóticas (inhibidores de GSK-3 beta), con inhibidores de la activación microglial (minociclina), antioxidantes (selegilina) o factores tróficos (IGF-1, hormona del crecimiento o eritropoyetina), que muestran efectos beneficiosos como tratamientos complementarios a la terapia antirretroviral (AU)


Neuroinflammation is a key process in the neuropathogenesis of AIDS virus since as a result of the aberrant activation of the chemokine receptors (CXCR4, CX3CR1 and CR5) produces proinflammatory cytokine release by infected cells, increases microglial neurotoxicity and generates lipoperoxides and reactive oxygen species (ROS) that eventually damage the neuron. Moreover, the neurotoxin Tat produces dendritic loss by interacting with the low-density lipoprotein receptor (LRP) and also overstimulates N-methyl D-aspartate receptors (NMDA). Furthermore, the aberrant interaction of glycoprotein gp120 with the CXCR4 chemokine receptor causes caspase-3-dependent apoptosis (ceramide is also released) activating apoptotic proteins (p53 and retinoblastoma), which are part of the neurotoxic mechanisms associated to neuronal dysfunction in neuroAIDS. Similarly, gliosis/microglial activation and the release of neurotoxic factors by infected monocytes with elevated amounts of certain chemokines in the cerebrospinal fluid (MCP-1 and fractalkine, among others) contribute to the neuropathogenesis of HIV-1. Alpha-synuclein and beta amyloid deposits have also been detected in post mortem brains of seropositives patients. In addition, there are studies have detected several systemic markers related with the degenerative effects of the virus and its neurotoxins on the central nervous system; such as osteopontin, CD163 and fractalkine, among others. Lastly, clinical trials have been conducted using protective strategies related that attempt to inhibit apoptotic proteins (GSK-3 beta), microglial activation inhibitors (minocycline), antioxidants (selegiline) or trophic factors (IGF-1, growth hormone or erythropoietin). These trials have shown that their treatments are beneficial and complementary to treat complications of HIV/AIDS (AU)


Humans , AIDS Dementia Complex/drug therapy , HIV Infections/complications , HIV Envelope Protein gp120/adverse effects , Chemokines, CX3C , Anti-Retroviral Agents/therapeutic use , Microglia
10.
Glia ; 58(13): 1630-9, 2010 Oct.
Article En | MEDLINE | ID: mdl-20578038

A number of human immunodeficiency virus type-1 (HIV) positive subjects are also opiate abusers. These individuals are at high risk to develop neurological complications. However, little is still known about the molecular mechanism(s) linking opiates and HIV neurotoxicity. To learn more, we exposed rat neuronal/glial cultures prepared from different brain areas to opiate agonists and HIV envelope glycoproteins gp120IIIB or BaL. These strains bind to CXCR4 and CCR5 chemokine receptors, respectively, and promote neuronal death. Morphine did not synergize the toxic effect of gp120IIIB but inhibited the cytotoxic property of gp120BaL. This effect was blocked by naloxone and reproduced by the mu opioid receptor agonist DAMGO. To examine the potential mechanism(s) of neuroprotection, we determined the effect of morphine on the release of chemokines CCL5 and CXCL12 in neurons, astrocytes, and microglia cultures. CCL5 has been shown to prevent gp120BaL neurotoxicity while CXCL12 decreases neuronal survival. Morphine elicited a time-dependent release of CCL5 but failed to affect the release of CXCL12. This effect was observed only in primary cultures of astrocytes. To examine the role of endogenous CCL5 in the neuroprotective activity of morphine, mixed cerebellar neurons/glial cells were immunoneutralized against CCL5 prior to morphine and gp120 treatment. In these cells the neuroprotective effect of opiate agonists was blocked. Our data suggest that morphine may exhibit a neuroprotective activity against M-tropic gp120 through the release of CCL5 from astrocytes.


Astrocytes/drug effects , Chemokine CCL5/metabolism , HIV Envelope Protein gp120/adverse effects , Morphine/pharmacology , Narcotics/pharmacology , Analgesics, Opioid/pharmacology , Animals , Animals, Newborn , Cell Survival/drug effects , Cells, Cultured , Cerebellum/cytology , Cytarabine/metabolism , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Enzyme-Linked Immunosorbent Assay/methods , Gene Expression Regulation/drug effects , Hippocampus/cytology , Neurons/drug effects , Neurons/metabolism , Rats , Rats, Sprague-Dawley
11.
J Neurosci Methods ; 185(2): 236-45, 2010 Jan 15.
Article En | MEDLINE | ID: mdl-19837113

Migraine is a neurovascular disorder that induces debilitating headaches associated with multiple symptoms including facial allodynia, characterized by heightened responsivity to normally innocuous mechanical stimuli. It is now well accepted that immune activation and immune-derived inflammatory mediators enhance pain responsivity, including the trigeminal system. Nociceptive ("pain" responsive) trigeminal nerves densely innervate the cranial meninges. We have recently proposed that the meninges may serve as a previously unidentified, key interface between the peripheral immune system and the CNS with potential implications for understanding underlying migraine mechanisms. Our focus here is the development of a model for facial allodynia associated with migraine. We developed a model wherein an indwelling catheter is placed between the skull and dura, allowing immunogenic stimuli to be administered over the dura in awake and freely moving rats. Since the catheter does not contact the brain itself, any proinflammatory cytokines induced following manipulation derive from resident or recruited meningeal immune cells. While surgery alone does not alter immune activation markers, TNF or IL6 mRNA and/or protein, it does decrease gene expression and increase protein expression of IL-1 at 4 days after surgery. Using this model we show the induction of facial allodynia in response to supradural administration of either the HIV glycoprotein gp120 or inflammatory soup (bradykinin, histamine, serotonin, and prostaglandin E2), and the induction of hindpaw allodynia in our model after inflammatory soup. This model allows time- and dose-dependent assessment of the relationship between changes in meningeal inflammation and corresponding exaggerated pain behaviors.


Disease Models, Animal , Face/physiopathology , Migraine Disorders/pathology , Migraine Disorders/physiopathology , Pain Threshold/physiology , Wakefulness , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, Differentiation, Myelomonocytic/metabolism , Blood-Brain Barrier/physiopathology , Bradykinin/adverse effects , CD11b Antigen/genetics , CD11b Antigen/metabolism , Catheters, Indwelling , Cytokines/genetics , Cytokines/metabolism , Dose-Response Relationship, Drug , Drug Combinations , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , HIV Envelope Protein gp120/adverse effects , Histamine/adverse effects , Hyperalgesia/etiology , Male , Migraine Disorders/chemically induced , Pain Measurement/methods , Rats , Rats, Sprague-Dawley , Serotonin/adverse effects , Time Factors
12.
J Neuropathol Exp Neurol ; 68(5): 456-73, 2009 May.
Article En | MEDLINE | ID: mdl-19525894

Human immunodeficiency virus 1 (HIV-1) encephalopathy is thought to result in part from the toxicity of HIV-1 envelope glycoprotein gp120 for neurons. Experimental systems for studying the effects of gp120 and other HIV proteins on the brain have been limited to the acute effects of recombinant proteins in vitro or in vivo in simian immunodeficiency virus-infected monkeys. We describe an experimental rodent model of ongoing gp120-induced neurotoxicity in which HIV-1 envelope is expressed in the brain using an SV40-derived gene delivery vector, SV(gp120). When it is inoculated stereotaxically into the rat caudate putamen, SV(gp120) caused a partly hemorrhagic lesion in which neuron and other cell apoptosis continues for at least 12 weeks. Human immunodeficiency virus gp120 is expressed throughout this time, and some apoptotic cells are gp120 positive. Malondialdehyde and 4-hydroxynonenal assays indicated that there was lipid peroxidation in these lesions. Prior administration of recombinant SV40 vectors carrying antioxidant enzymes, copper/ zinc superoxide dismutase or glutathione peroxidase, was protective against SV(gp120)-induced oxidative injury and apoptosis. Thus, in vivo inoculation of SV(gp120) into the rat caudate putamen causes ongoing oxidative stress and apoptosis in neurons and may therefore represent a useful animal model for studying the pathogenesis and treatment of HIV-1 envelope-related brain damage.


AIDS Dementia Complex/etiology , Disease Models, Animal , HIV Envelope Protein gp120/adverse effects , HIV Envelope Protein gp120/metabolism , HIV-1/pathogenicity , AIDS Dementia Complex/genetics , Adenoviruses, Simian , Aldehydes/metabolism , Animals , Calcium-Binding Proteins , Cell Death , DNA-Binding Proteins/metabolism , Female , Gene Expression Regulation, Viral/physiology , Genetic Vectors/administration & dosage , Glial Fibrillary Acidic Protein/metabolism , Glutathione Peroxidase/metabolism , Humans , In Situ Nick-End Labeling , Indoles , Microfilament Proteins , Neurons/pathology , Neurons/virology , Phosphopyruvate Hydratase/metabolism , Putamen/virology , Rats , Rats, Nude , Rats, Sprague-Dawley , Statistics, Nonparametric , Time Factors , Transduction, Genetic/methods
13.
Gene Ther ; 14(23): 1650-61, 2007 Dec.
Article En | MEDLINE | ID: mdl-17914406

Human immunodeficiency virus-1 (HIV-1) is the most frequent cause of dementia in adults under 40. We sought to use gene delivery to protect from HIV-1-related neuron loss. Because HIV-1 envelope (Env) gp120 elicits oxidant stress and apoptosis in cultured neurons, we established reproducible parameters of Env-mediated neurotoxicity in vivo, then tested neuroprotection using gene delivery of antioxidant enzymes. We injected 100-500 ng mul(-1)gp120 stereotaxically into rat caudate-putamens (CP) and assayed brains for apoptosis by terminal deoxynucleotidyl transferase-mediated biotinylated UTP nick end labeling (TUNEL) 6-h to 14-day post-injection. Peak apoptosis occurred 1 day after injection of 250 and 500 ng microl(-1)gp120. TUNEL-positive cells mostly expressed neuronal markers (NeuroTrace), although some expressed CD68 and so were most likely microglial cells. Finally, we compared neuroprotection from gp120-induced apoptosis provided by localized and generalized intra-central nervous system (CNS) gene delivery. Recombinant SV40 vectors carrying Cu/Zn superoxide dismutase (SOD1) or glutathione peroxidase (GPx1) were injected into the CP, where gp120 was administered 4-24 weeks later. Alternatively, we inoculated the vector into the lateral ventricle (LV), with or without prior intraperitoneal (i.p.) administration of mannitol. Intracerebral injection of SV(SOD1) or SV(GPx1) significantly protected neurons from gp120-induced apoptosis throughout the 24-week study. Intraventricular vector administration protected from gp120 neurotoxicity comparably, particularly if preceded by mannitol i.p. Thus, HIV-1 gp120 is neurotoxic in vivo, and intracerebral or intra-ventricular administration of rSV40 vectors carrying antioxidant enzymes is neuroprotective. These findings suggest the potential utility of both localized and widespread gene delivery in treating neuroAIDS and other CNS diseases characterized by excessive oxidative stress.


AIDS Dementia Complex/prevention & control , Antioxidants/therapeutic use , Genetic Therapy/methods , HIV Envelope Protein gp120/adverse effects , HIV-1 , Neurons/virology , AIDS Dementia Complex/virology , Animals , Apoptosis , Brain/metabolism , Brain/virology , Female , Genetic Vectors/administration & dosage , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Humans , Injections , Injections, Intraventricular , Models, Animal , Neurons/metabolism , Oxidative Stress , Rats , Rats, Sprague-Dawley , Simian virus 40/genetics , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Transduction, Genetic/methods
14.
J Acquir Immune Defic Syndr ; 46(1): 48-55, 2007 Sep 01.
Article En | MEDLINE | ID: mdl-17909315

BACKGROUND: The development of an effective HIV-1 vaccine is critical to control the pandemic. A prime-boost HIV-1 vaccine trial assessing safety and immunogenicity was conducted in Thailand as part of an evaluation of candidate regimens for a phase 3 efficacy trial. METHODS: ALVAC-HIV (vCP1521), expressing circulating recombinant form 01_AE (CRF01_AE) gp120/subtype B LAI and subtype B Gag/Protease boosted with recombinant envelope oligomeric CRF01_AE gp160 (ogp160) or bivalent CRF01_AE/subtype B gp120 CM235/SF2, was evaluated in a phase 1/II trial of 130 HIV-negative Thai adults. RESULTS: One hundred forty volunteers were enrolled, and 130 completed all safety and immunogenicity visits. Reactogenicity was common but generally mild, and there was no significant difference in the adverse event rate between vaccine and placebo recipients (P = 0.26). There were 7 serious adverse events during the follow-up period, none of which were vaccine related. Cumulative HIV-specific, CD8-mediated, cytotoxic T-lymphocyte responses were observed in 11 (25%) of 44 subjects who received ALVAC boosted by bivalent gp120 and in 5 (11%) of 45 subjects who received ALVAC boosted by ogp160, but these differences were not statistically significant compared with those in placebo recipients (P = 0.62 and P = 0.37, respectively). HIV-specific lymphoproliferative responses were detected in 84% of subunit-boosted vaccine recipients and in 10% of placebo recipients. Neutralizing antibody responses to CRF01_AE and subtype B laboratory strains were seen in 95% of ogp160-boosted and 100% of gp120 B/E-boosted vaccinees, respectively. CONCLUSIONS: These 2 different prime-boost regimens seem to be safe and displayed cell-mediated immune responses consistent with those in other trials of canarypox vectors.


AIDS Vaccines/immunology , HIV Envelope Protein gp120/immunology , HIV Envelope Protein gp160/immunology , AIDS Vaccines/administration & dosage , AIDS Vaccines/adverse effects , Adult , Cell Proliferation , Double-Blind Method , Female , HIV Antibodies/immunology , HIV Antigens/administration & dosage , HIV Antigens/adverse effects , HIV Antigens/immunology , HIV Envelope Protein gp120/administration & dosage , HIV Envelope Protein gp120/adverse effects , HIV Envelope Protein gp160/administration & dosage , HIV Envelope Protein gp160/adverse effects , HIV Infections/immunology , HIV Infections/prevention & control , Humans , Lymphocytes/immunology , Male , Middle Aged , Protein Binding , Vaccination
15.
Neurosci Lett ; 396(1): 50-3, 2006 Mar 20.
Article En | MEDLINE | ID: mdl-16343773

Human immunodeficiency virus (HIV)-wasting syndrome might be facilitated by the HIVgp120 affecting the immunological system. We studied the effect (subchronic administration: 5 days) of HIVgp120, and a few immune-response mediators: regulated upon activation normal T-cell expressed and presumably secreted (RANTES), stromal derived factor-1alpha (SDF-1alpha), macrophage-derived chemokine (MDC), and their combination, on food and water intake in rats, motor control and pain perception. Eighty male adult Wistar rats received an intracerebroventricular (icv) administration of: vehicle 5 microl/day or 0.92 nmol daily of HIVgp120IIIB, RANTES, SDF-1alpha, or MDC, and the combination of RANTES+HIVgp120IIIB, SDF-1alpha+HIVgp120IIIB, or MDC+HIVgp120IIIB. Food and water intake was measured every day during administration, and 24 and 48 h after the last administration. Rats were also weighed the first and the last day of experiment in order to detect the impact of these treatments in the body weight. HIVgp120IIIB significantly decreased food and water intake. These rats gain less weight than the control (vehicle) and chemokines-treated subjects with exception of those treated with SDF-1alpha that also gain less weight. In addition, HIVgp120 deteriorated motor control. HIVgp120IIIB effects on food and water intake, and motor control were prevented by these chemokines. HIVgp120+RANTES, HIVgp120+SDF-1alpha, and SDF-1alpha alone induced hyperalgesia. Results suggest an interaction between HIVgp120 and the chemokine system to generate the HIV-wasting syndrome, the motor abnormalities and changes in pain perception.


Appetite Regulation/immunology , Chemokine CCL5/immunology , Chemokines, CC/immunology , Chemokines, CXC/immunology , Drinking/immunology , HIV Envelope Protein gp120/immunology , Animals , Appetite Regulation/drug effects , Body Weight/drug effects , Body Weight/immunology , Chemokine CCL22 , Chemokine CCL5/pharmacology , Chemokine CXCL12 , Chemokines, CC/pharmacology , Chemokines, CXC/pharmacology , Drinking/drug effects , Drug Administration Schedule , Drug Therapy, Combination , HIV Envelope Protein gp120/adverse effects , HIV Wasting Syndrome/immunology , HIV Wasting Syndrome/physiopathology , HIV Wasting Syndrome/virology , HIV-1/immunology , Male , Movement Disorders/immunology , Movement Disorders/virology , Pain/chemically induced , Pain/immunology , Pain/virology , Rats
16.
J Acquir Immune Defic Syndr ; 29(3): 254-61, 2002 Mar 01.
Article En | MEDLINE | ID: mdl-11873074

To test the safety and immunogenicity of a high-titered preparation of ALVAC-HIV vCP205 in both high-risk and low-risk persons and to evaluate variations in dosing schedule, we conducted a multicenter, randomized, double-blind trial of this vector in combination with recombinant subunit gp120 in 150 HIV-1-seronegative volunteers. The high-titered ALVAC vaccine was well tolerated; adverse events were minimal and not influenced by dosing. At day 728, the cumulative probability of a cytotoxic T-lymphocyte (CTL) response was 76% (95% confidence interval [CI]: 64%-89%) among volunteers receiving vaccine, and the net amount attributable to vaccination was 50% (CI: 16%; 74%). The net probability of a repeated positive CTL response by day 728 was 50% (CI: 21%; 64%). There was a significant difference in CTL response at day 182 between volunteers who had received four doses versus three doses of vCP205 (42% vs. 24%, p =.052). The CTL response was similar in high-risk volunteers and vaccinia-naive volunteers compared with vaccinia-immune volunteers. Neutralizing antibody responses were detected in 95% of vaccinees at day 287, with higher geometric mean titers in recipients of sequential versus simultaneous dosing of the two vaccines and in vaccinia-naive volunteers. This high-titered preparation of ALVAC-HIV vCP205 in combination with gp120 was safe and immunogenic in a diverse group of HIV-1-seronegative volunteers.


AIDS Vaccines/adverse effects , AIDS Vaccines/immunology , HIV Envelope Protein gp120/immunology , AIDS Vaccines/genetics , Adult , Double-Blind Method , Female , HIV Antibodies/blood , HIV Envelope Protein gp120/adverse effects , HIV Envelope Protein gp120/genetics , HIV Infections/diagnosis , HIV Seronegativity/immunology , HIV-1/immunology , Humans , Immunization Schedule , Male , Neutralization Tests , Recombinant Proteins/adverse effects , Recombinant Proteins/genetics , Recombinant Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Combined , Vaccines, Synthetic
17.
Rev Invest Clin ; 54(5): 437-52, 2002.
Article Es | MEDLINE | ID: mdl-12587419

The AIDS or HIV associated dementia is a cognitive-motor disease, characterized by a strong deficit of several cognitive processes such as attention, memory, sensory perception, motor control among others. The HIV associated dementia affects 30% of adult to 50% of infant HIV positive subjects. Since neurons are not infected by HIV, its principal target in the brain is microglia. The pathophysiology of this syndrome, therefore, remains to be disclosed. Several hypothesis have been proposed, one of them suggests that opportunistic infections can affect the brain. Another hypothesis suggests that microglia secretes toxic products as a result of HIV infection and those are the ones causing the damage and finally, the hypothesis, suggesting that the brain is damaged as a result of the insult caused by HIV-derived proteins. In vitro studies suggest that the HIVgp120, a viral surface protein, is highly neurotoxic. For example HIVgp120 increases cytoplasmic Ca+2 by two ways: facilitating glutamate neurotransmission increasing Ca+2 conductance, and activating the IP3 pathway, facilitating Ca+2 release from the smooth endoplasmic reticulum. This Ca+2 in turn, activates several internal signaling pathways such as the MAPK pathway. We use an animal model to test the HIVgp120 effect on neurophysiological signals and behavior as well as several pharmacological approaches to prevent the HIVgp120 neurotoxic effects. This review updates with the most recent literature discussing the potential mechanisms implicated in the pathophysiology of the AIDS dementia complex. We, in addition, hope the reader will be able to correlate the clinical symptoms observed in the HIV infected subjects and the HIVgp120-induced behavioral changes observed in animal models. Likewise, we discuss the new drugs we are testing, in order to offer a new pharmacological treatment to the patient.


AIDS Dementia Complex/etiology , HIV Envelope Protein gp120/adverse effects , HIV-1/physiology , Microglia/pathology , AIDS Dementia Complex/pathology , AIDS Dementia Complex/physiopathology , AIDS-Related Opportunistic Infections/complications , Animals , Anti-HIV Agents/pharmacology , Calcium Signaling , Cell Death , Chemokines/metabolism , Disease Models, Animal , Drug Design , Event-Related Potentials, P300 , Glutamic Acid/physiology , HIV Envelope Protein gp120/physiology , Haplorhini , Humans , MAP Kinase Signaling System , Prion Diseases/pathology , Prion Diseases/veterinary , Prion Diseases/virology , Receptors, Chemokine/physiology , Receptors, Virus/physiology , Simian Acquired Immunodeficiency Syndrome/pathology , Virus Replication
18.
Vaccine ; 19(23-24): 3033-42, 2001 Apr 30.
Article En | MEDLINE | ID: mdl-11311997

Thirty-three HIV-seronegative adults were recruited into a Phase I safety and immunogenicity HIV-1 vaccine trial. The immunogens were as follows: a synthetic, monovalent, octameric HIV-1 MN V3 peptide in aluminum hydroxide (alum) adjuvant administered by intramuscular delivery; and a similar product encapsulated in biodegradable micro-spheres composed of co-polymers of lactic and glycolic acids, administered by the oral route. These were administered in three sequential oral doses, followed by a parenteral boost. No serious adverse experiences were observed. Oral administration of this vaccine, alone or in combination with parenteral boosting, resulted in no significant humoral, cellular, or mucosal immune responses.


AIDS Vaccines/administration & dosage , HIV Envelope Protein gp120/administration & dosage , Peptide Fragments/administration & dosage , AIDS Vaccines/adverse effects , AIDS Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Administration, Oral , Adult , Aluminum Hydroxide/administration & dosage , Antibody-Producing Cells/immunology , Female , HIV Antibodies/biosynthesis , HIV Envelope Protein gp120/adverse effects , HIV Envelope Protein gp120/immunology , HIV Seronegativity/immunology , HIV-1/immunology , Humans , Immunity, Cellular , Immunity, Mucosal , Immunization, Secondary , In Vitro Techniques , Lymphocyte Activation , Male , Microspheres , Middle Aged , Peptide Fragments/adverse effects , Peptide Fragments/immunology , Safety , T-Lymphocytes, Cytotoxic/immunology
19.
Brain Res ; 861(1): 105-16, 2000 Apr 07.
Article En | MEDLINE | ID: mdl-10751570

Astrocytes and microglia in the spinal cord have recently been reported to contribute to the development of peripheral inflammation-induced exaggerated pain states. Both lowering of thermal pain threshold (thermal hyperalgesia) and lowering of response threshold to light tactile stimuli (mechanical allodynia) have been reported. The notion that spinal cord glia are potential mediators of such effects is based on the disruption of these exaggerated pain states by drugs thought to preferentially affect glial function. Activation of astrocytes and microglia can release many of the same substances that are known to mediate thermal hyperalgesia and mechanical allodynia. The aim of the present series of studies was to determine whether exaggerated pain states could also be created in rats by direct, intraspinal immune activation of astrocytes and microglia. The immune stimulus used was peri-spinal (intrathecal, i.t.) application of the Human Immunodeficiency Virus type 1 (HIV-1) envelope glycoprotein, gp120. This portion of HIV-1 is known to bind to and activate microglia and astrocytes. Robust thermal hyperalgesia (tail-flick, TF, and Hargreaves tests) and mechanical allodynia (von Frey and touch-evoked agitation tests) were observed in response to i.t. gp120. Heat denaturing of the complex protein structure of gp120 blocked gp120-induced thermal hyperalgesia. Lastly, both thermal hyperalgesia and mechanical allodynia to i.t. gp120 were blocked by spinal pretreatment with drugs (fluorocitrate and CNI-1493) thought to preferentially disrupt glial function.


HIV Envelope Protein gp120/adverse effects , Hyperalgesia/chemically induced , Neuroglia/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Citrates/therapeutic use , Hot Temperature/adverse effects , Hydrazones/therapeutic use , Hyperalgesia/drug therapy , Male , Rats , Rats, Sprague-Dawley , Specific Pathogen-Free Organisms , Spinal Cord/drug effects , Touch/drug effects
20.
Vaccine ; 18(13): 1166-77, 2000 Jan 18.
Article En | MEDLINE | ID: mdl-10649617

Thirty healthy HIV negative volunteers were randomised to receive 200 micrograms of rgp120W61D in either: 3D-MPL and QS21, with an oil and water emulsion (SBAS-2) (13); or 3D-MPL and QS21 (SBAS-1) (11); or alum (six). Immunizations were given at 0, 4 and 28 weeks and 23 (77%) participants completed the schedule. Adverse events were more frequent (P < 0.001) and more severe (P < 0.001) in the SBAS-2 group. Binding antibodies to the homologous rgp120W61D were detected after the first immunisation only in those receiving SBAS-1 and SBAS-2, were maximal after the third immunization in all three groups, and persisted to week 84 only in the novel adjuvant groups. These differences were significant (p = 0.02). Neutralising antibodies to TCLA-strains of HIV-1 were observed after the second immunization in all three groups, were maximal after the third immunization, but did not neutralise homologous or heterologous PBMC derived primary HIV-1 isolates. Proliferative T-cell responses to rgp120W61D were maximal after the second immunization and reached very high values in the SBAS-2 group. HIV-1 specific CD8+ MHC Class I restricted cytotoxic T-lymphocytes were not seen in a subset of participants tested at a single timepoint. SBAS-2 with rgp120W61D induced antibody titres as high as those seen in HIV infection, but the quality of the antibodies remained different in that there was no evidence of primary isolate neutralisation. Although cell-mediated immunity was enhanced by SBAS-2 in terms of lymphoproliferative responses, HIV-1 specific CD8+ cytotoxicity was not demonstrated.


AIDS Vaccines/immunology , Adjuvants, Immunologic/pharmacology , HIV Envelope Protein gp120/immunology , HIV Seronegativity/immunology , HIV-1/immunology , Lipid A/analogs & derivatives , Vaccines, Synthetic/immunology , AIDS Vaccines/adverse effects , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/adverse effects , Adult , Female , Follow-Up Studies , HIV Antibodies/blood , HIV Antibodies/metabolism , HIV Envelope Protein gp120/adverse effects , HIV Envelope Protein gp120/metabolism , Humans , Immunization Schedule , Lipid A/administration & dosage , Lipid A/adverse effects , Lipid A/pharmacology , Male , Neutralization Tests , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/adverse effects
...