Allopregnanolone and neuroHIV

Buy Lab Tests Online

madman

Super Moderator
Abstract

Forty years into the HIV pandemic, approximately 50% of infected individuals still suffer from a constellation of neurological disorders collectively known as ‘neuroHIV.’
Although combination antiretroviral therapy (cART) has been a tremendous success, in its present form, it cannot eradicate HIV. Reservoirs of virus reside within the central nervous system, serving as sources of HIV virotoxins that damage mitochondria and promote neurotoxicity. Although understudied, there is evidence that HIV or the HIV regulatory protein, trans-activator of transcription (Tat), can dysregulate neurosteroid formation potentially contributing to endocrine dysfunction. People living with HIV commonly suffer from endocrine disorders, including hypercortisolemia accompanied by paradoxical adrenal insufficiency upon stress. Age-related comorbidities often onset sooner and with greater magnitude among people living with HIV and are commonly accompanied by hypogonadism. In the post-cART era, these derangements of the hypothalamic-pituitary-adrenal and -gonadal axes are secondary (i.e., relegated to the brain) and indicative of neuroendocrine dysfunction. We review the clinical and preclinical evidence for neuroendocrine dysfunction in HIV, the capacity for hormone therapeutics to play an ameliorative role, and the future steroid-based therapeutics that may have efficacy as novel adjunctives to cART.




1 | INTRODUCTION


It has been 40 years since the beginning of the HIV pandemic and an estimated 37.6 million people continue to live with the virus worldwide.1 The tremendous success of combination antiretroviral therapy (cART) has increased the life expectancies of people living with HIV (PLWH) and has greatly improved morbidity. However, the transformation of HIV into a chronic care illness has also revealed the long-term consequences of living with the infection. Even in the post-cART era, many individuals continue to suffer from neurological symptoms including an increased prevalence of cognitive impairment, major depression, generalized anxiety disorder, neuropathic pain, and motor dysfunction, collectively referred to as “neuroHIV”.2 Identifying and treating the mechanisms of these neurological deficits is the subject of intense investigation. To this end, the influence of neuroHIV on the neuroendocrine system has become increasingly apparent. In this review, we summarize the recent advances made in our understanding of HIV effects on neuroendocrine function and the potential benefits of steroid-based therapeutics, including the progesterone metabolite, 5α-pregnan3α-ol-20-one (3α,5α-THP or allopregnanolone), on neuroHIV and HIV viremia. Taken together, the data support a reciprocal relationship between HIV and neuroendocrine status, in which HIV dysregulates the neuroendocrine axes, facilitating stress- and age-related comorbidities that neuroendocrine-based therapeutics may help ameliorate.




2 | HIV-MEDIATED NEUROLOGICAL DYSFUNCTION


In the post-cART era, approximately 50% of PLWH suffer from neuroHIV.3 Although cART has reduced the incidence of the most severe form of neurocognitive impairment, HIV-associated dementia (present in ~20% of HIV patients in the pre-cART era vs. ~2% in the post-cART era),3 the proportion of individuals that continue to express any cognitive impairment has remained stable, albeit the symptoms are markedly milder.

2.1 | The HIV central nervous system reservoir

2.2 | Mechanisms of neuroHIV




3 | STEROIDOGENIC DYSREGULATION BY HIV

The endocrine system has long been known to be perturbed by HIV infection with patients presenting with dysfunction of the adrenals, gonads, pituitary, and thyroid.65 However, it is becoming apparent that the interactions between HIV and the endocrine system are dynamic. Endogenous steroids influence HIV replication and neuropathology and, conversely, HIV virotoxins can influence steroid formation. As such, the relationship between neuroendocrine function and neuroHIV symptomatology is reciprocal.

3.1 | Steroid hormones modulate HIV replication

3.2 | Steroid hormones ameliorate HIV-related neuropathology

3.3 | Pregnane steroids ameliorate HIV Tat-induced behavioral pathology


3.4 | HIV disrupts central and circulating steroidogenesis




4 | HIV EFFECTS ON HYPOTHALAMIC-PITUITARY-ADRENAL AND-GONADAL AXES

HIV can exert a profound influence on circulating steroid hormone production. These effects can reduce circulating steroid content via actions at endocrine glands, such as the adrenals or gonads (i.e. primary insufficiency), or via actions targeted to the source of steroid promoting corticotropins and gonadotropins in the hypothalamus and anterior pituitary (i.e. secondary insufficiency). The latter is far more common in the post-cART era, emphasizing the need for HIV therapeutics with efficacy in the CNS. As a result, PLWH is commonly affected by disruptions to the hypothalamic-pituitary-adrenal (HPA) and –gonadal (HPG) axes.91,92 Such dysfunction likely contributes to the neuropsychiatric components of neuroHIV and the age-related comorbidities that are observed earlier in life and to a greater magnitude among PLWH.

4.1 | HPA stress axis dysregulation in people living with HIV

4.2 | Potential mechanisms involved in HIV dysregulation of the HPA axis

4.3 | HPG stress axis dysregulation in people living with HIV


4.4 | Potential mechanisms involved in HIV dysregulation of the HPG axis




5 | STEROID-BASED THERAPEUTICS FOR THE TREATMENT OF HIV


5.1 | Novel adjunct therapeutics for HIV suppression

Antiretroviral therapeutics have dramatically increased the life expectancy of PLWH. However, these drugs are not able to eradicate the virus, nor neuroHIV, given that they cannot target reservoirs such as those within the CNS and do not target certain virotoxins such as Tat. It is notable that several promising leads are based on a steroid scaffold. Most notably, didehydro-cortistatin A (dCA), an analog of a steroid-like alkaloid obtained from marine sponge, is demonstrated in vitro to bind to the Tat-TAR complex, blocking HIV replication without producing cellular toxicity, to attenuate HIV-mediated cytokine expression, and to inhibit behavioral effects promoted by Tat in vivo. 185,186 dCA was found to selectively bind the unstructured basic region of Tat.187 A combination of dCA and cART suppressed active HIV viral replication, reactivation, and rebound of the latent viral reservoir.188 Additional chemical derivatives of dCA were sought to rationalize their ability to dock at specific binding sites of the Tat protein.187 Given the ability of dCA to inhibit Tat expression during early viral replication and penetrate latent viral reservoirs in the brain with good bioavailability, dCA and its novel steroidal-based analogs hold potential as future cART adjuncts.187-189

Additional estrogen-based therapeutics have also been identified, including selective estrogen receptor β agonists, such as (S)- equol and several phytoestrogens. (S)-equol improved sensorimotor gating and motivational deficits in HIV transgenic rats190-192 and prevented combined cocaine and HIV-mediated synaptopathy.193 The phytoestrogens, daidzein, and liquiritigenin, restored Tat-mediated synaptodendritic recovery.194 Our own work has focused on the potential therapeutic advantages of neurosteroids.

Neurosteroids are synthesized de novo in the brain from cholesterol195 and can also reach the brain from peripheral sources such as the adrenals and gonads.196 AlloP is perhaps the most well-characterized neurosteroid with a pharmacodynamic profile that is suitable for potentially offsetting the effects of HIV Tat (Figure 1). Given the excitotoxic profile exerted by Tat, the actions of AlloP as a potent positive allosteric modulator of GABAA receptors are expected to reinstate excitatory-inhibitory balance via the influx of Cl−. Moreover, AlloP may act as an antagonist of L-type Ca2+ channels,197,198 further attenuating the excitotoxic actions of Tat. We have seen promising evidence for AlloP to offset Tat-mediated neurotoxicity in vitro (Figure 2) and to attenuate Tat-mediated behavioral interactions with opioids in vivo. 81 Although AlloP is a small molecule that readily crosses the blood-brain barrier and is well-tolerated,199 it is also rapidly re-distributed from the brain, accumulates in adipose tissue, and has a short elimination half-life.200,201 As such, we are currently working to synthesize AlloP analogs with anti-Tat and anti-viremic properties. A ligand structural alignment of dCA, (S)- Equol, and AlloP reveals structural similarities that may help explain their anti-HIV activities (Figure 6). For example, both terminal ends of the molecules contain polar elements (oxygen or a nitrogen atom). Induced-fit docking of dCA in the NMR-derived structure of Tat (Protein Data Bank: 1K5K) reveals important hydrogen bonding interactions between dCA and the R49 and R52 residues, which are part of the identified motif (the ARM domain) (Figure 7). Induced-fit docking of AlloP in this binding site also reveals important hydrogen bonding interactions between AlloP and the R49 residue, in addition to strong hydrophobic interactions (Figure 7). These preliminary data suggest that dCA and AlloP may target a shared Tat binding site that may partly underlie their potential anti-Tat efficacy. In light of this, the development of novel AlloP analogs may hold promise with respect to potential future cART adjunctive therapeutics.
 

Attachments

  • 2021OCT19-ALLO-NEUROHIV-jne.13047.pdf
    1.6 MB · Views: 40
Defy Medical TRT clinic doctor

madman

Super Moderator
FIGURE 1 Mechanisms of direct neuronal damage for the HIV proteins, trans-activator of transcription (Tat), and glycoprotein 120 (gp120). HIV proteins can directly drive intracellular Ca2+ (partly by activation of NMDA receptors, L-type Ca2+ channels, or activation of chemokine receptor-mediated signaling) or can indirectly activate NMDA receptors via activation of low-density lipoprotein receptor-related protein (LRP). The downstream effects of HIV proteins dysregulate mitochondrial membrane potential, drive the formation of reactive oxygen species (ROS), and promote cell injury and death (left). Allopregnanolone is a potent positive allosteric modulator of GABAA receptors that can antagonize L-type Ca2+ channels and restore mitochondrial homeostasis, potentially off-setting the excitotoxic actions of HIV proteins. Allopregnanolone-sulfate is an antagonist of NMDA receptors (right).
Screenshot (8510).png
 

madman

Super Moderator
FIGURE 2 HIV Tat (50–100 nm) increases intracellular calcium, depolarizes mitochondria, and promotes microgliosis and/or neurotoxicity in murine striatal medium spiny neurons (A), murine microglia (B), or differentiated human SH-SY5Y neuroblastoma cells (C). Pretreatment with allopregnanolone (AlloP; 100 nm) attenuates Tat-mediated effects. *Significant increase from control following Tat exposure. †Significant AlloPmediated rescue from Tat exposure.
Screenshot (8511).png
 

madman

Super Moderator
FIGURE 3 Expression of HIV Tat protein in mice increases basal corticotropin-releasing hormone (CRH) and corticosterone. Upon stress, male (but not female) mice demonstrate adrenal insufficiency. The effects of Tat to dysregulate cholesterol homeostasis and alter steroidogenesis may contribute to the dysregulation of the hypothalamic-pituitary–adrenal (HPA) axis. In normative systems, neuroactive steroids such as allopregnanolone restore HPA axis homeostasis, downregulating CRH, adrenocorticotropic hormone (ACTH), and glucocorticoid production
Screenshot (8512).png
 

madman

Super Moderator
FIGURE 4 Compared to Tat(−) controls, male Tat(+) mice demonstrate greater corticosterone at baseline with paradoxical adrenal insufficiency in response to a stressor (15-min forced swim) or pharmacological inhibition of the glucocorticoid receptor (GR) via RU-486 (A). Compared to controls, female Tat(+) mice also demonstrate increased corticosterone at baseline; however, no differences are observed following 15-min forced swim stress or administration of RU-486 (B). *Significant difference from Tat(−) control46-48
Screenshot (8513).png
 

madman

Super Moderator
FIGURE 5 Middle-aged (16–19 months old) female Tat(+) mice had greater learning deficits on a radial arm water maze than their Tat(−) counterparts, irrespective of estropausal status (A). Anxiety-like behavior on an elevated plus-maze was greater among peri- and postestropausal mice compared to those that were pre-estropausal, irrespective of Tat exposure (B). Male or female Tat(+) mice had reduced grip strength compared to Tat(−) controls. Middle-aged males (11–13 months old) had reduced grip strength compared to young adult males (6–8 months old) (C). *Significant difference from Tat(−) control. †Significant difference from young adult male or pre-estropausal female group177
Screenshot (8514).png
 

madman

Super Moderator
FIGURE 6 Didehydro-cortistatin A (orange), allopregnanolone (cyan), and (S)-equol (pink) demonstrate structural similarities (A) that are evidenced in a ligand-structure alignment (B)
Screenshot (8515).png
 

madman

Super Moderator
FIGURE 7 Molecular docking of didehydro-cortistatin A (dCA) (orange) and allopregnanolone (cyan) in HIV Tat (Protein Data Bank: 1K5K). Tat is shown in a surface representation in the zoom-out view (A) and in a ribbon representation in the zoom-in view (B, C). Key residues of Tat for ligand binding are shown in stick representation
Screenshot (8516).png
 
Buy Lab Tests Online
Defy Medical TRT clinic

Sponsors

enclomiphene
nelson vergel coaching for men
Discounted Labs
TRT in UK Balance my hormones
Testosterone books nelson vergel
Register on ExcelMale.com
Trimix HCG Offer Excelmale
Thumos USA men's mentoring and coaching
Testosterone TRT HRT Doctor Near Me

Online statistics

Members online
1
Guests online
7
Total visitors
8

Latest posts

bodybuilder test discounted labs
Top