Molecular Mimicry and Functional Convergence: A Unifying Hypothesis for the Pathophysiology of Long COVID Based on the HIV Proteome
Long COVID may mimic HIV’s chronic effects via molecular mimicry and functional convergence, with SARS-CoV-2 proteins triggering neuro, cardio, and immune dysfunction through persistent virotoxins.
Introduction
The Clinical Enigma of Long COVID (PASC)
The global health landscape has been irrevocably altered by the COVID-19 pandemic, caused by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). While the acute phase of the disease has been the primary focus of clinical and research efforts, a second, more insidious crisis has emerged in its wake: Post-Acute Sequelae of SARS-CoV-2 infection (PASC), colloquially known as Long COVID.1 PASC is a complex, multi-systemic condition characterized by a constellation of symptoms that persist or newly appear weeks to months after the initial infection has resolved.4 The clinical presentation of PASC is remarkably heterogeneous, affecting a wide range of organ systems. Commonly reported symptom clusters include profound fatigue, post-exertional malaise, and significant neurocognitive impairment, often described as "brain fog".7 Cardiovascular complications are also prevalent, encompassing myocarditis, dysautonomia, endothelial dysfunction, and thrombotic events.10 Furthermore, a state of persistent immune dysregulation appears to be a central feature, linking many of these disparate clinical manifestations.9
This condition affects a substantial portion of COVID-19 survivors, with some estimates suggesting that up to 30% of individuals experience persistent symptoms, regardless of the severity of their initial illness.5 The profound impact on quality of life and the potential for long-term disability represent a looming public health crisis.1 A primary challenge in addressing PASC is the absence of a unified pathophysiological model that can account for its diverse and seemingly disconnected symptoms.7 The leading hypotheses—including viral persistence, autoimmunity, reactivation of latent viruses, and microbiome dysbiosis—each explain certain facets of the disease but often fail to provide a comprehensive, integrated framework.14 This diagnostic and mechanistic uncertainty severely hampers the development of effective, targeted therapies.
Introducing the HIV Mimicry/Convergence Hypothesis
This report advances a central thesis: that the complex and chronic pathology of PASC can be mechanistically illuminated by examining the molecular mimicry and functional convergence between SARS-CoV-2 proteins and the well-characterized pathogenic proteins of the Human Immunodeficiency Virus (HIV). While SARS-CoV-2 and HIV belong to different viral families and employ distinct replication strategies, a growing body of evidence suggests they have evolved analogous molecular tools to manipulate the host immune system, invade tissues, and establish chronic pathology.17 This convergence of function results in strikingly similar long-term clinical sequelae, particularly in the neurocognitive, cardiovascular, and immunological domains.
The study of HIV-associated neurocognitive disorders (HAND), HIV-related cardiovascular disease, and the chronic immune activation characteristic of AIDS has spanned over four decades, yielding a deep and granular understanding of how specific viral proteins (such as gp120, Tat, Nef, Vpr, and Vpu) drive long-term, multi-systemic disease. This extensive body of knowledge provides a robust, pre-existing conceptual framework—a pathogenic playbook—that can be leveraged to dissect the mechanisms of PASC.21 By mapping the functions of SARS-CoV-2 proteins onto their HIV counterparts, it becomes possible to generate testable hypotheses that connect specific molecular interactions to the clinical manifestations of Long COVID. This approach posits that SARS-CoV-2, in its evolutionary interaction with the human host, has stumbled upon pathogenic strategies long perfected by HIV, a master of chronic infection and immune evasion.
Roadmap of the Report and Summary Table
This report will systematically explore the evidence supporting this hypothesis. It will begin by establishing the foundational principles of molecular mimicry and convergent viral evolution. Subsequently, it will conduct a detailed comparative analysis of the SARS-CoV-2 Spike glycoprotein and HIV gp120 as central virotoxins driving neuroinflammation and endothelial damage. The analysis will then extend to the functional analogs of HIV's accessory and regulatory proteins (Tat, Nef, Vpr, Vpu) within the SARS-CoV-2 proteome, linking their roles to specific PASC pathologies. Finally, the report will synthesize these findings into a unified, multi-hit model of PASC, place this model in the context of the broader scientific debate, and explore its profound implications for diagnostics, risk stratification, and the repurposing of therapies from the extensive HIV armamentarium. The following table provides a high-level summary of the core protein comparisons that form the basis of this report.
Table 1: Functional Convergence and Molecular Mimicry between HIV-1 and SARS-CoV-2 Proteins
Section 1: The Foundations of Viral Mimicry and Convergent Pathogenesis
1.1. Molecular Mimicry: The Immunological Basis for Post-Infectious Autoimmunity
Molecular mimicry is a well-established immunological phenomenon wherein structural similarities between pathogen-derived antigens and host self-antigens can precipitate a breakdown in self-tolerance, leading to autoimmunity.22 The immune system, upon encountering a pathogen, mounts a robust response to eliminate the foreign invader. However, if a component of that pathogen—for instance, a short peptide sequence—closely resembles a sequence in a human protein, the antibodies or T cells generated against the pathogen may cross-react with the host's own tissues.22 This cross-reactivity can initiate an autoimmune cascade, resulting in chronic inflammation and tissue damage long after the initial infection has been cleared. This mechanism is considered a primary driver of many post-infectious autoimmune diseases.22
This mimicry can manifest at different structural levels. The most commonly studied form involves short, linear amino acid sequences (e.g., pentapeptides or heptapeptides) that are identical or highly similar between a viral protein and a human protein.23 These short mimics are sufficient to be presented by major histocompatibility complex (MHC) molecules and recognized by T cells, or to be targeted by antibodies, thereby triggering a cross-reactive response.23 Beyond linear sequences, mimicry can also occur at the level of three-dimensional protein structures, where disparate primary sequences fold into similar shapes that can be recognized by the immune system.29 Viruses frequently employ this strategy to evade the host immune system; by resembling "self," they can partially hide from immune surveillance.23 However, this strategy is a double-edged sword. While it may offer the virus a temporary advantage, it risks awakening autoreactive immune cells that can cause lasting harm to the host. The "molecular mimicry trade-off hypothesis" posits that viruses must carefully balance the immune-evasive benefits of mimicry against potential functional compromises to their proteins, which may explain the evolutionary prevalence of short, epitope-sized mimics that minimize this cost.25
The development of post-infectious autoimmunity is often conceptualized within the framework of the "autoimmune disease triangle," which requires three components: a triggering pathogen, a hyperactive or dysregulated immune system, and a genetically susceptible host.26 SARS-CoV-2 infection perfectly fulfills the first two criteria, as the virus itself is the trigger and it is known to induce a profound state of immune hyper-activation, or "cytokine storm".26 This framework has been used to explain the link between various viruses and specific autoimmune conditions, such as the association between Epstein-Barr virus (EBV) and Multiple Sclerosis (MS), or Parvovirus B19 and Systemic Lupus Erythematosus (SLE).22 The emergence of a wide spectrum of autoimmune phenomena following COVID-19 strongly suggests that molecular mimicry is a key pathogenic mechanism at play.26
1.2. Convergent Evolution in Disparate RNA Viruses: The Case of SARS-CoV-2 and HIV-1
Beyond the direct sharing of amino acid sequences, SARS-CoV-2 and HIV-1 exhibit a remarkable degree of functional convergence—the independent evolution of similar biological traits to solve common adaptive challenges.31 Despite their origins in different viral families (Coronaviridae and Retroviridae, respectively), both are zoonotic RNA viruses that successfully made the leap to humans and caused devastating pandemics.19 In adapting to the same host environment—the human body and its complex immune system—both viruses appear to have arrived at analogous solutions for infection, propagation, and persistence.
This convergence is evident across multiple stages of the viral life cycle and pathogenesis:
Host Cell Entry: Both viruses have evolved sophisticated biophysical strategies to engage the host cell surface. They initially recognize and bind to lipid raft microdomains, which act as "landing strips".31 They then modulate the electrostatic potential of their surface glycoproteins (the positively charged Spike NTD/RBD for SARS-CoV-2 and V3 loop for HIV-1) to facilitate "surfing" across the electronegative cell surface in search of their primary protein receptors.31 This shared strategy underscores a fundamental convergence in the physics of viral attachment. Functionally, the SARS-CoV-2 Spike S1 subunit and HIV-1 gp120 are analogs for receptor binding, while the Spike S2 subunit and gp41 are analogs for mediating membrane fusion.31
Systemic Inflammation: A hallmark of severe disease in both infections is the induction of a massive, dysregulated inflammatory response, or "cytokine storm".17 This hyperinflammation is a key driver of acute organ damage and is increasingly recognized as a precursor to the chronic inflammatory state that underlies long-term complications, such as the elevated risk of cardiovascular disease seen in both PASC and HIV-infected individuals.17
Immune Dysregulation: Both SARS-CoV-2 and HIV-1 infections are characterized by a profound disruption of the adaptive immune system. A key shared feature is the induction of T-lymphocyte deficiency (lymphopenia), particularly of CD4+ T cells.18 In the chronic phase, both can lead to a state of T-cell exhaustion, marked by the sustained upregulation of inhibitory receptors like Programmed cell death protein 1 (PD-1), which impairs the ability of T cells to effectively control the virus and contributes to persistent immune dysfunction.18
Downstream Pathologies: The convergence in molecular and immunological mechanisms leads to a convergence in clinical outcomes. Both infections are associated with gastrointestinal disturbances, a wide range of neurological symptoms, and a heightened risk of thrombotic complications, which may be driven by shared mechanisms like the aberrant formation of neutrophil extracellular traps (NETs).17
The consistent pattern of convergent evolution between these two disparate viruses is not merely an academic curiosity; it provides a powerful predictive framework. The long-term pathogenic pathways of HIV, which have been meticulously studied for decades, can serve as a detailed roadmap for understanding the chronic pathologies of PASC. Because similar selective pressures from the host immune system have driven the evolution of functionally analogous viral proteins, it is logical to infer that the downstream consequences of these protein functions will also be similar. This perspective elevates the extensive body of neuroHIV and cardio-HIV research from a source of interesting parallels to a directly applicable, hypothesis-generating engine for investigating PASC, allowing researchers to anticipate pathogenic mechanisms and identify potential therapeutic targets based on a well-established model of chronic viral disease.
Section 2: The Envelope Glycoproteins—Spike and gp120 as Central Virotoxins
2.1. Structural Homologies and Functional Parallels
The primary interface between both SARS-CoV-2 and HIV-1 and their host is their respective envelope glycoproteins: the Spike (S) protein and the glycoprotein 120 (gp120). Early in the pandemic, a controversial and since-withdrawn preprint posited an "uncanny similarity" between the two, based on the identification of four short amino acid insertions in the Spike protein that showed identity to sequences in HIV-1 gp120 and the Gag polyprotein.34 This claim was met with significant scientific skepticism, as the identified inserts were extremely short, and such matches could easily occur by chance in vast protein sequence databases.
While the notion of direct, significant sequence homology remains contentious, the evidence for functional and structural convergence is far more compelling and scientifically robust. Both Spike and gp120 are considered potent virotoxins, capable of inducing cellular damage and inflammation independent of productive viral infection.31 They share several key characteristics that underscore their analogous roles. Both are trimeric structures that stud the viral surface and are heavily glycosylated, forming a "glycan shield" that helps the virus evade recognition by host antibodies.33 Their primary function is to mediate viral entry, but their pathogenic roles extend far beyond this initial step. A critical point of convergence lies in their evolutionary dynamics. The Receptor Binding Domain (RBD) and N-terminal domain (NTD) of the SARS-CoV-2 Spike protein and the V3 loop of HIV-1 gp120 are functionally analogous, rapidly evolving regions.31 Both viruses have been shown to accumulate mutations in these specific domains that increase their net positive surface electrostatic potential. This biophysical change enhances their initial, non-specific binding to negatively charged components of the host cell membrane (like gangliosides in lipid rafts), thereby increasing the efficiency of infection and providing a kinetic advantage.31 Crucially, this same evolutionary trajectory toward increased electropositivity also confers stronger resistance to neutralizing antibodies, representing a shared strategy for immune escape.31
2.2. Mechanisms of Neuroinflammation and Glial Cell Activation
A direct causal link can be drawn from the virotoxic properties of Spike and gp120 to the debilitating neurological symptoms of PASC, such as cognitive deficits and "brain fog".8 The mechanisms by which these two distinct proteins induce a state of chronic neuroinflammation are remarkably similar, providing a strong basis for the parallel neurocognitive syndromes observed in PASC (neuro-PASC) and HIV-associated neurocognitive disorder (HAND).21
The SARS-CoV-2 Spike protein has been shown to be neuroinvasive. Evidence indicates that the protein, and not necessarily the intact virus, can cross the blood-brain barrier (BBB) and has been found to persist in the brain parenchyma, meninges, and even the bone marrow of the skull for months or years following acute infection.41 Once in the central nervous system (CNS), the Spike protein acts as a potent inflammatory stimulus, functioning as a pathogen-associated molecular pattern (PAMP).44 It directly activates the brain's resident immune cells, microglia, and perivascular mast cells, primarily through pattern recognition receptors such as Toll-like receptor 4 (TLR4).44 This activation triggers a classic neuroinflammatory cascade, leading to the release of a barrage of pro-inflammatory cytokines and chemokines, including interleukin-1β (IL-1β), IL-6, and tumor necrosis factor-α (TNF-α).44 This sustained inflammatory milieu leads to synaptic remodeling, excitotoxicity, and ultimately, neuronal damage and dysfunction.
This pathway is a striking parallel to the well-documented neurotoxicity of HIV's gp120. For decades, research has established gp120 as a primary driver of HAND.49 Shed from infected cells, soluble gp120 activates microglia, which then release IL-1β.49 This cytokine, in turn, acts directly on neurons, altering synaptic function by increasing the number of inhibitory synapses and potentiating the function of N-methyl-D-aspartate (NMDA) receptors, a key mechanism of excitotoxic neuronal injury.49 Furthermore, both gp120 and the HIV Tat protein contribute to the breakdown of the BBB by inducing oxidative stress in brain endothelial cells and disrupting the tight junction proteins that maintain the barrier's integrity.51 The convergence is clear: in both diseases, a viral envelope glycoprotein gains access to the CNS, directly triggers glial cells to produce a similar profile of inflammatory cytokines, and disrupts the BBB, culminating in a state of chronic neuroinflammation that underlies cognitive impairment.
2.3. Endothelial Dysfunction and Cardiovascular Pathology
The Spike and gp120 proteins are also central to the cardiovascular complications seen in PASC and chronic HIV infection, including endothelial dysfunction, a prothrombotic state, and myocardial inflammation.10 The vascular endothelium is a primary target in both diseases.
SARS-CoV-2 infection is increasingly understood as a pan-vascular disease, with endothelial injury (endotheliitis) as a core pathological feature.10 The Spike protein initiates this damage by binding to angiotensin-converting enzyme 2 (ACE2) receptors, which are highly expressed on endothelial cells.54 This interaction leads to direct cell injury, inflammation, and a critical shift in endothelial function towards a pro-coagulant and pro-inflammatory state.53 This endothelial dysfunction is a key driver of the microthrombosis and hypercoagulability that characterize severe COVID-19 and are suspected to contribute significantly to the multi-organ symptoms of PASC.12 Furthermore, molecular mimicry between the Spike protein and cardiac self-antigens, such as α-myosin, has been proposed as a plausible mechanism for the myocarditis observed both after infection and, rarely, after vaccination.58
This pathogenic pathway mirrors the effects of HIV's gp120 on the vasculature. Soluble gp120 is known to directly damage endothelial cells, inducing apoptosis and dysregulating the production of angiogenic factors.60 This chronic endothelial injury is a major contributor to the significantly elevated risk of atherosclerosis, myocardial infarction, and other cardiovascular diseases observed in people living with HIV (PLWH), even those with suppressed viral loads.60 Both viruses, through their functionally analogous envelope glycoproteins, trigger a common cascade of endothelial activation, inflammation, and coagulation system dysregulation.53 The elevation of Vascular Endothelial Growth Factor A (VEGF-A) in both infections is a notable shared pathway, further linking the two to pathological angiogenesis and vascular dysfunction.63
2.4. Systemic Immune Dysregulation and Autoimmunity
Beyond localized tissue damage, the persistent presence of these viral glycoproteins drives a state of systemic and long-lasting immune dysregulation that is central to the chronic nature of both PASC and HIV/AIDS. The detection of Spike protein fragments in the blood and tissues of PASC patients long after the acute phase suggests a source of chronic antigenic stimulation.9 This persistent exposure can lead to T-cell exhaustion, a state of cellular dysfunction characterized by reduced effector function and sustained expression of inhibitory receptors, which is a hallmark of unresolved chronic viral infections like HIV.9
This is functionally analogous to the role of soluble gp120 in HIV pathogenesis. Shed from virions and infected cells, sgp120 circulates as a "pan-toxin," broadly activating the immune system by inducing cytokine production from monocytes, macrophages, and T cells.66 It also coats uninfected bystander CD4+ T cells, marking them for destruction by antibody-dependent cellular cytotoxicity (ADCC). This contributes to the progressive depletion of CD4+ T cells and fuels a state of chronic immune activation and inflammation that persists even when viral replication is controlled by antiretroviral therapy (ART).66
This chronic immune stimulation, coupled with molecular mimicry, provides a fertile ground for the development of autoimmunity. The SARS-CoV-2 Spike protein has been found to share numerous short peptide sequences (pentapeptides) with a wide array of human proteins.28 Critically, some of these shared sequences are found in proteins essential for nervous system function, such as myelin-associated glycoprotein and other components of axon and myelin homeostasis.70 An immune response targeting these sequences on the Spike protein could cross-react with these neural proteins, potentially triggering autoimmune demyelinating syndromes or other neurological disorders observed in PASC. This parallels findings in HIV, where the gp120 protein has been shown to mimic regions of human T-cell receptors (TCRs).71 This mimicry can lead to the generation of lymphocytotoxic autoantibodies that attack the host's own T cells, contributing to the profound immunodeficiency that defines AIDS.71 In both cases, the envelope glycoprotein serves as the nexus linking viral infection to a subsequent autoimmune attack on the host, providing a direct mechanistic explanation for the autoimmune features of PASC and HIV.27
The long-term persistence of the SARS-CoV-2 Spike protein or its fragments in various body tissues, including immunoprivileged sites like the brain, represents a crucial parallel to the persistence of soluble gp120 in HIV.41 This "Virotoxin Persistence" hypothesis proposes a unifying mechanism that can account for the primary triad of PASC pathologies. The continued presence of this foreign, biologically active protein, even in the absence of active viral replication, acts as a chronic inflammatory and antigenic trigger. This single driver can explain the seemingly disparate symptoms of PASC: it fuels neuroinflammation through the direct activation of glial cells in the CNS; it perpetuates cardiovascular damage via sustained endothelial inflammation and dysfunction; and it drives systemic immune dysregulation by causing T-cell exhaustion and providing the antigenic stimulus for autoimmunity through molecular mimicry. This model moves beyond general concepts like "chronic inflammation" to propose a specific, actionable driver—the persistent virotoxin—that connects the leading hypotheses of PASC into a cohesive pathogenic framework.
Section 3: Functional Analogs of HIV Accessory and Regulatory Proteins in SARS-CoV-2 Pathogenesis
3.1. Tat-like Effects: Neurotoxicity and Angiogenic Mimicry
The HIV trans-activator of transcription (Tat) protein is a small regulatory protein essential for viral replication, but its pathogenic functions extend far beyond this role. Tat is secreted from infected cells and can be taken up by neighboring uninfected cells, where it acts as a potent toxin, particularly in the CNS.73 The neurotoxicity of Tat is multifaceted, contributing significantly to HAND through several mechanisms: it induces excitotoxicity by interacting with NMDA receptors, promotes oxidative stress, causes mitochondrial dysfunction, and disrupts neuronal cytoskeletal dynamics by triggering the hyper-activation of cyclin-dependent kinase 5 (CDK5).75 Beyond its neurotoxic effects, Tat exhibits a remarkable form of functional mimicry; it acts as an angiogenic factor by binding directly to the Vascular Endothelial Growth Factor Receptor 2 (VEGFR-2), the primary receptor for VEGF-A. This interaction promotes pathological angiogenesis and vascular permeability, contributing to cardiovascular complications and Kaposi's sarcoma.63
While the SARS-CoV-2 genome does not encode a direct structural or sequence homolog of Tat, the pathogenic outcomes of Tat's activity are strikingly mirrored in severe COVID-19 and PASC. The profound neuroinflammatory state driven by the persistent Spike protein and the associated systemic cytokine storm can induce secondary neurotoxicity through pathways that overlap with those triggered by Tat, such as oxidative stress and excitotoxicity.47 Furthermore, a key point of convergence is the dysregulation of the VEGF pathway. Both severe COVID-19 and chronic HIV infection are characterized by significantly elevated levels of circulating VEGF-A.63 In COVID-19, this is likely driven by hypoxia and the inflammatory response, whereas in HIV, it is driven by both inflammation and the direct action of Tat. Regardless of the upstream trigger, the downstream consequence is the same: a shared pathway of pathological angiogenesis, endothelial dysfunction, and increased vascular permeability. This demonstrates a clear functional convergence where different viral strategies culminate in the same end-organ pathology.
3.2. Nef-like Functions: Hijacking Host Pathways for Immune Evasion
The HIV Negative factor (Nef) protein is a small accessory protein that is indispensable for high-titer viral replication and pathogenesis in vivo.83 Nef is a quintessential viral manipulator; although it lacks any intrinsic enzymatic activity, it functions by hijacking a multitude of host cell protein trafficking and signaling pathways.83 Its most well-characterized functions are aimed at creating a favorable environment for the virus by subverting the host immune response. Nef downregulates key cell surface receptors from infected cells, including CD4 (to prevent superinfection and ADCC) and MHC class I molecules (to hide the infected cell from cytotoxic T lymphocytes).83 It also enhances the intrinsic infectivity of viral particles by counteracting the host restriction factor SERINC5.83 In the CNS, Nef contributes to neuroinflammation by dysregulating astrocyte function and compromising the integrity of the BBB.75
SARS-CoV-2 has evolved its own distinct set of accessory proteins that exhibit functionally analogous, Nef-like roles in immune manipulation. The SARS-CoV-2 proteome contains several open reading frame (ORF) proteins that are not part of the final virion structure but are crucial for antagonizing host defenses. For example, the ORF8 protein has been shown to be a structural and functional mimic of the host cytokine IL-17.87 By binding to the IL-17 receptor, ORF8 can activate downstream inflammatory signaling pathways, contributing to the cytokine storm and immune pathogenesis of severe COVID-19.87 Other accessory proteins, such as ORF7a and ORF9b, have also been implicated in modulating host immune responses, including the suppression of innate immune signaling.88 This demonstrates a convergent evolutionary strategy: both viruses have developed a specialized toolkit of non-structural proteins dedicated to hijacking host pathways, manipulating the immune environment, and facilitating viral persistence, which in turn contributes to the chronic inflammation seen in both PASC and HIV.
3.3. Vpr-like Cardiotoxicity and Systemic Inflammation
Viral protein R (Vpr) is another of HIV's multifunctional accessory proteins. It is known to play roles in the nuclear import of the viral pre-integration complex and in inducing G2/M cell cycle arrest and apoptosis in infected T cells.89 Beyond these roles, compelling evidence from animal models has established Vpr as a direct cardiotoxin. Transgenic mice expressing Vpr specifically in cardiomyocytes develop a severe cardiac pathology characterized by atrial cardiomyocyte mitosis, cardiac dysrhythmia, chamber dilation, and eventual congestive heart failure.91 Further studies have shown that circulating Vpr can induce senescence and fibrosis in heart tissue.93 These findings establish a direct, causal link between a specific HIV protein and the development of cardiomyopathy, a known complication of long-term HIV infection.89
While a specific Vpr mimic has not been identified in the SARS-CoV-2 proteome, the functional outcome of direct cardiac injury is a prominent feature of both severe acute COVID-19 and cardiovascular PASC.94 Myocardial injury, myocarditis, and arrhythmias are well-documented complications. The SARS-CoV-2 Spike protein itself has been implicated in direct cardiac damage, and CCL2-mediated endothelial injury has been shown to drive cardiac dysfunction in PASC models.95 The convergence here lies in the shared phenotype of virus-associated cardiac pathology. While the specific viral protein may differ, both HIV (via Vpr) and SARS-CoV-2 (via Spike and systemic inflammation) have the capacity to inflict direct or indirect damage on the heart muscle, leading to similar clinical manifestations of cardiomyopathy and heart failure. This highlights a shared vulnerability of the cardiovascular system to the pathogenic effects of these two distinct viruses.
3.4. Vpu-like Antagonism of Innate Immunity and Inflammasome Activation
Viral protein U (Vpu) is an accessory protein unique to HIV-1 and some related simian immunodeficiency viruses (SIVs).96 It performs two canonical and critical functions that promote viral dissemination. First, it mediates the degradation of newly synthesized CD4 molecules in the endoplasmic reticulum, preventing them from interfering with the viral envelope protein during assembly.97 Second, and perhaps more importantly, it counteracts a key host innate immune restriction factor known as tetherin (also called BST-2), which would otherwise trap newly formed virions on the surface of the infected cell, preventing their release.98 More recent research has uncovered a more subtle but profound immunomodulatory role for Vpu: it acts as a potent inhibitor of both the canonical and non-canonical NF-κB signaling pathways.100 It achieves this by hijacking the SCFβ-TrCP ubiquitin ligase complex, thereby preventing the degradation of IκB and dampening the host's antiviral and pro-inflammatory gene expression program late in the viral lifecycle.100 Concurrently, various HIV proteins, including Vpu, have been shown to be potent triggers of the NLRP3 inflammasome, a key platform for the production of inflammatory cytokines IL-1β and IL-18.102
This dual ability to both suppress certain antiviral pathways (NF-κB) while activating others (the inflammasome) highlights a sophisticated viral strategy to sculpt the immune environment. SARS-CoV-2 has convergently evolved its own mechanisms to achieve similar goals. Numerous SARS-CoV-2 proteins are known to antagonize the host's innate immune response, particularly the type I interferon system, which is crucial for establishing a successful infection.88 At the same time, the hyperinflammatory state of severe COVID-19 is inextricably linked to the massive activation of inflammasomes, particularly NLRP3, which drives the cytokine storm and pyroptotic cell death.47 This comparison reveals that both viruses have developed mechanisms to dysregulate the same central hubs of innate immunity and inflammation. They must suppress initial antiviral defenses to gain a foothold, but their components can later trigger an excessive inflammatory response that, while damaging to the host, may facilitate viral spread and contribute to the chronic pathologies seen in PASC and AIDS.
The existence of a diverse suite of accessory proteins in both HIV (Nef, Vpr, Vpu, Vif) and SARS-CoV-2 (ORF3a, ORF6, ORF7a, ORF8, etc.) points to a convergent evolutionary principle for chronic viral pathogenesis. A successful chronic pathogen requires not only the basic structural proteins for building new virions but also a specialized "toolkit" of accessory proteins designed to systematically dismantle host defenses, modulate inflammation, hijack cellular machinery, and ensure long-term persistence. While the specific proteins in the SARS-CoV-2 toolkit are not homologous to those of HIV, they appear to perform functionally analogous roles. This "Accessory Protein Toolkit" model suggests that the remarkable heterogeneity of PASC symptoms may be explained by individual differences in the expression, persistence, and combination of these various SARS-CoV-2 "tools." For instance, a patient with a persistent, Nef-like immune-evasive protein might present with immunological exhaustion, while another with a Vpu-like inflammatory trigger might exhibit signs of hyperinflammation and autoimmunity. This framework shifts the focus from a single causative agent to the combinatorial pathogenic effects of a persistent viral toolkit.
Section 4: A Unified Pathophysiological Model and the Broader Scientific Context
4.1. Synthesizing the Evidence for a Multi-Hit Hypothesis
The evidence presented across the preceding sections, detailing the functional convergence between SARS-CoV-2 and HIV proteins, allows for the synthesis of a unified, multi-hit pathophysiological model for PASC. This model posits that Long COVID is not a monolithic entity driven by a single mechanism but is rather the result of the cumulative and often synergistic effects of multiple SARS-CoV-2 proteins that mimic the distinct pathogenic functions of different HIV proteins. The specific clinical phenotype, or symptom cluster, that manifests in an individual patient is likely determined by the unique combination of these "hits," the persistence of the specific viral proteins involved, and the individual's genetic and immunological background.
This model provides a powerful framework for explaining the profound heterogeneity of PASC. For example:
Neuro-PASC: The debilitating "brain fog," memory loss, and cognitive deficits could be the result of a "double hit." The first hit is the direct neuroinflammatory effect of the persistent Spike protein, acting in a gp120-like manner to activate microglia and disrupt the BBB. The second hit comes from the systemic inflammation and vascular damage that can cause secondary neurotoxicity through excitotoxic and oxidative stress pathways, a functional parallel to the effects of HIV Tat.
Cardiovascular PASC: The array of cardiovascular complications, including myocarditis, endothelial dysfunction, and thrombotic events, can be understood as a multi-hit process. The primary hit is the gp120-like function of the Spike protein, causing direct endotheliitis and promoting a pro-coagulant state. This may be compounded by a Vpr-like effect from other viral components causing direct cardiotoxicity, and systemic inflammation further exacerbating vascular damage.
Immunological PASC: The profound fatigue, post-exertional malaise, and autoimmune phenomena can be explained by the convergence of several hits on the immune system. The chronic antigenic stimulation from persistent Spike protein drives T-cell exhaustion, a common outcome in both diseases. This is likely combined with specific immune evasion tactics from accessory proteins performing Nef-like functions and a dysregulation of innate inflammatory pathways, such as the inflammasome, mirroring the effects of Vpu.
This multi-hit model, grounded in the established pathogenic principles of HIV, successfully integrates the disparate clinical features of PASC into a cohesive whole. It moves beyond single-cause explanations to embrace the complexity of the syndrome, offering a more nuanced and mechanistically plausible foundation for future research and clinical management.
4.2. The Scientific Debate and Alternative Hypotheses
The HIV mimicry and functional convergence hypothesis provides a compelling explanatory framework, particularly for the multi-systemic nature of PASC. Its primary strength lies in its ability to leverage decades of research into a well-understood chronic viral illness to make sense of a new one. However, it is essential to critically evaluate this hypothesis and situate it within the broader scientific discourse on PASC pathogenesis. A key limitation is that, with the exception of some short peptide sequences, direct sequence homology between the respective proteins is often weak or non-existent.33 Therefore, the argument rests heavily on the concept of functional convergence, which, while powerful, can be more challenging to prove definitively than direct mimicry.
It is also crucial to recognize that this hypothesis is not mutually exclusive with other leading theories of PASC; in fact, it often provides a deeper mechanistic underpinning for them.17 The main alternative or complementary hypotheses include:
Viral Persistence: This theory, supported by findings of SARS-CoV-2 RNA and protein in tissues like the gut and brain months after infection, is a cornerstone of PASC research.14 The HIV mimicry/convergence hypothesis does not contradict this; rather, it provides a detailed explanation for
how this persistent viral material causes ongoing pathology through the specific virotoxic and immunomodulatory functions of the remaining proteins.Reactivation of Latent Viruses: A state of immune dysregulation induced by COVID-19 can lead to the reactivation of latent herpesviruses, such as Epstein-Barr virus (EBV) and Human Herpesvirus 6 (HHV-6), which are themselves known triggers of chronic fatigue-like syndromes.5 The functional convergence model accommodates this by positing that SARS-CoV-2 proteins, acting like their HIV counterparts, create the very state of immune exhaustion and dysregulation that allows these latent viruses to re-emerge.
Autoimmunity: The de novo production of autoantibodies is a well-documented feature of PASC.106 The molecular mimicry component of the central hypothesis provides the most direct and well-established mechanism for how a viral infection can trigger this break in self-tolerance.27
Microbiome Dysbiosis: Significant alterations in the gut microbiome are strongly correlated with PASC symptoms and systemic inflammation.14 This dysbiosis can be viewed as both a consequence of the initial viral infection and the subsequent systemic inflammation driven by persistent viral proteins, and a contributing factor that perpetuates the inflammatory state.
Endothelial Dysfunction and Microclots: The persistence of endothelial damage and the formation of amyloid-containing microclots are considered by many to be a central pathological driver of PASC, explaining symptoms from brain fog to cardiovascular issues.10 The HIV convergence hypothesis directly addresses this through the gp120/Spike virotoxin mechanism, which identifies the viral glycoprotein as the primary instigator of the endotheliitis that initiates this cascade.
Ultimately, PASC is likely a heterogeneous syndrome with multiple overlapping pathogenic pathways. The strength of the HIV functional convergence model is its ability to serve as an integrating framework that connects these disparate phenomena—viral persistence, autoimmunity, and endothelial damage—through the specific, well-understood actions of viral proteins.
Section 5: Clinical and Therapeutic Implications
5.1. Toward Novel Diagnostics and Risk Stratification
The application of the HIV mimicry and functional convergence framework has the potential to revolutionize the clinical approach to PASC. Currently, Long COVID is often treated as a single, albeit heterogeneous, diagnosis based on a collection of symptoms. This framework allows for a paradigm shift toward mechanism-based diagnostics and risk stratification, enabling a more precise, personalized approach to patient care.
Instead of a general PASC diagnosis, it may become possible to phenotype patients based on the dominant underlying pathogenic pathway suggested by the HIV model. For instance:
Neurological PASC Phenotyping: Patients presenting with prominent "brain fog" and cognitive decline could be screened for biomarkers of specific pathways. The presence of high-titer autoantibodies that cross-react with both Spike protein and myelin-associated proteins would suggest a "Spike-driven autoimmune neuro-PASC" phenotype, warranting close neurological monitoring and potentially targeted immunotherapies.
Cardiovascular PASC Phenotyping: Individuals with persistent cardiovascular symptoms could be assessed for markers of ongoing endothelial activation (e.g., soluble VCAM-1, von Willebrand factor) and microclot formation. Persistently elevated levels would point toward a "gp120-like vascular PASC" phenotype, flagging them for aggressive cardiovascular risk management and potential anti-thrombotic or endothelial-stabilizing therapies.
Immunological PASC Phenotyping: Deep immune profiling could identify patients with a dominant "Nef-like" T-cell exhaustion profile (e.g., high PD-1 expression on SARS-CoV-2-specific T cells) versus those with a "Vpu-like" hyperinflammatory profile characterized by elevated inflammasome-related cytokines. This distinction could guide the choice between immunostimulatory and immunosuppressive therapeutic strategies.
This approach would transform PASC from a descriptive syndrome into a set of distinct endotypes, each with a defined biological signature, allowing for more accurate prognostication and the development of targeted interventions.
5.2. Repurposing Therapeutics from the HIV Playbook
Perhaps the most immediate and impactful implication of the functional convergence hypothesis is the rationale it provides for repurposing therapeutics from the well-stocked HIV drug development pipeline to treat PASC. If SARS-CoV-2 proteins are causing disease through mechanisms analogous to HIV proteins, then drugs designed to block those HIV-specific pathways may prove effective in PASC.
Repurposing Antivirals: This logic is the explicit basis for ongoing clinical trials investigating HIV antivirals for Long COVID.109
Maraviroc (Selzentry): This drug is a CCR5 antagonist, blocking one of the key co-receptors HIV uses for entry. In the context of PASC, its therapeutic potential lies not in blocking viral entry, but in its ability to modulate the activity of monocytes and macrophages, which express CCR5 and are key drivers of chronic inflammation. By blocking CCR5 signaling, Maraviroc may be able to dampen the persistent inflammation that contributes to vascular and neurological damage.109
Tenofovir/Emtricitabine (Truvada): This combination drug is a reverse transcriptase inhibitor. Its rationale for use in PASC is twofold. First, some in vitro studies have suggested it has activity against SARS-CoV-2.109 Second, and perhaps more relevant to PASC, it is a potent inhibitor of EBV replication.109 Given that EBV reactivation is a leading hypothesis for PASC symptoms, Truvada could work by suppressing this secondary viral driver.
Repurposing Immunomodulators: The chronic immune activation and inflammation central to PASC are reminiscent of the pathologies seen in PLWH. Consequently, immunomodulatory strategies that have been explored or used to manage HIV-associated inflammation are highly relevant for PASC. This includes Janus kinase (JAK) inhibitors like baricitinib, which can broadly dampen cytokine signaling, and more targeted cytokine inhibitors, such as anti-IL-6 (tocilizumab) or anti-TNF-α agents.112 Clinical trials are actively investigating these agents for PASC, implicitly testing the hypothesis that a shared inflammatory phenotype warrants a shared therapeutic approach.113
5.3. Future Research Directions
The HIV mimicry and functional convergence hypothesis, while compelling, requires rigorous scientific validation. The framework itself generates a clear and actionable roadmap for future research that could definitively establish its validity and translate it into clinical practice. Key research priorities should include:
Mechanistic Validation of Virotoxin Persistence:
Can advanced proteomics and imaging techniques definitively identify and quantify persistent SARS-CoV-2 protein fragments (particularly from Spike, ORF8, and other accessory proteins) in tissue biopsies (e.g., gut, lymph node) and cerebrospinal fluid from PASC patients?
Can these isolated protein fragments be shown to have direct pathological activity in relevant in vitro human cell systems (e.g., inducing cytokine release from primary microglia, causing permeability changes in brain endothelial cell models, or activating platelets)?
Defining the Autoimmune Landscape:
3. Can high-throughput serological assays be developed to systematically screen PASC patient cohorts for autoantibodies that cross-react with both specific SARS-CoV-2 proteins and their putative human protein mimics (e.g., Spike and α-myosin; Spike and myelin-associated glycoprotein)?
4. Do the presence and titer of such cross-reactive autoantibodies correlate with specific clinical phenotypes (e.g., myocarditis, demyelinating syndromes)?Immunophenotyping and Biomarker Discovery:
5. Can deep immune profiling (e.g., single-cell RNA sequencing, mass cytometry) of PASC patients identify distinct immunophenotypes that align with the proposed functional mimicry pathways (e.g., a "Nef-like" T-cell exhaustion signature vs. a "Vpu-like" hyperinflammatory signature)?
6. Can these immunophenotypes be correlated with specific symptom clusters and used to develop biomarkers for patient stratification?Translational and Clinical Trial Validation:
7. In ongoing and future clinical trials of repurposed HIV drugs (e.g., Maraviroc) for PASC, can mechanism-based biomarkers be incorporated to test the hypothesis directly? For example, does treatment with Maraviroc lead to a measurable reduction in activated monocyte populations and associated inflammatory markers in patients who show clinical improvement?
By systematically addressing these questions, the research community can move to either validate, refine, or refute this hypothesis. If validated, this framework holds the potential to transform PASC from a mystifying collection of symptoms into a set of well-defined, mechanistically-driven syndromes with clear and logical targets for therapeutic intervention, leveraging decades of hard-won knowledge from the fight against HIV.
Acknowledgement
I acknowledge the assistance of Gemini AI in the preparation of the subject research plan, the execution of the research, and the preparation of this report.
Works cited
Pathophysiological, immunological, and inflammatory features of long COVID - PMC, accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10932978/
Exploring the Pathophysiology of Long COVID: The Central Role of Low-Grade Inflammation and Multisystem Involvement - PMC, accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11204317/
Long COVID in people living with HIV - NATAP, accessed August 18, 2025, https://www.natap.org/2023/HIV/long_covid_in_people_living_with_hiv.4.pdf
Long COVID in people living with HIV - PMC - PubMed Central, accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10167544/
Long COVID or Post-acute Sequelae of COVID-19 (PASC): An Overview of Biological Factors That May Contribute to Persistent Symptoms - Frontiers, accessed August 18, 2025, https://www.frontiersin.org/journals/microbiology/articles/10.3389/fmicb.2021.698169/full
Pathogenic mechanisms of post-acute sequelae of SARS-CoV-2 infection (PASC) - eLife, accessed August 18, 2025, https://elifesciences.org/articles/86002
Long COVID-19 Pathophysiology: What Do We Know So Far? - MDPI, accessed August 18, 2025, https://www.mdpi.com/2076-2607/11/10/2458
Long COVID Brain Fog: What It Is and How to Manage It > News > Yale Medicine, accessed August 18, 2025, https://www.yalemedicine.org/news/how-to-manage-long-covid-brain-fog
In Patients with Long COVID, Immune Cells Don't Follow the Rules, accessed August 18, 2025, https://gladstone.org/news/patients-long-covid-immune-cells-dont-follow-rules
Role of Endothelium in Cardiovascular Sequelae of Long COVID - MDPI, accessed August 18, 2025, https://www.mdpi.com/2227-9059/11/8/2239
Vascular dysfunction in COVID-19 patients: update on SARS-CoV-2 infection of endothelial cells and the role of long non-coding RNAs - Portland Press, accessed August 18, 2025, https://portlandpress.com/clinsci/article/136/21/1571/232077/Vascular-dysfunction-in-COVID-19-patients-update
The cross-talk of lung and heart complications in COVID-19: Endothelial cells dysfunction, thrombosis, and treatment - Frontiers, accessed August 18, 2025, https://www.frontiersin.org/journals/cardiovascular-medicine/articles/10.3389/fcvm.2022.957006/full
Immune mechanisms underlying COVID-19 pathology and post ..., accessed August 18, 2025, https://elifesciences.org/articles/86014
Persistent SARS-CoV-2 Infection, EBV, HHV-6 and Other Factors ..., accessed August 18, 2025, https://www.mdpi.com/1999-4915/15/2/400
Biological mechanisms underpinning the development of long COVID - PMC, accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10193768/
Long COVID: A proposed hypothesis-driven model of viral persistence for the pathophysiology of the syndrome | Request PDF - ResearchGate, accessed August 18, 2025, https://www.researchgate.net/publication/360448050_Long_COVID_A_proposed_hypothesis-driven_model_of_viral_persistence_for_the_pathophysiology_of_the_syndrome
On Similarities and Differences of HIV and SARS Cov-2; Open Questions Relevant for COVID-19 Disease - Fortune Journals, accessed August 18, 2025, https://www.fortunejournals.com/articles/on-similarities-and-differences-of-hiv-and-sars-cov2-open-questions-relevant-for-covid19-disease.html
SARS-CoV-2 and HIV-1: So Different yet so Alike. Immune ..., accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9608044/
Similarities and differences between HIV and SARS-CoV-2, accessed August 18, 2025, https://www.medsci.org/v18p0846.htm
Similarities and differences between HIV and SARS-CoV-2 - PMC, accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7797543/
Studying the neuropsychological sequelae of SARS-CoV-2: lessons ..., accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7471564/
Molecular mimicry, hyperactive immune system, and SARS-COV-2 are three prerequisites of the autoimmune disease triangle following COVID-19 infection - PubMed Central, accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9393178/
Molecular mimicry between SARS-CoV-2 and human proteins - ResearchGate, accessed August 18, 2025, https://www.researchgate.net/publication/349490095_Molecular_mimicry_between_SARS-CoV-2_and_human_proteins
Molecular mimicry and autoimmunity in the time of COVID-19 - PMC - PubMed Central, accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10258587/
Molecular Mimicry as a Mechanism of Viral Immune Evasion and Autoimmunity - PMC, accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10942439/
Molecular mimicry, hyperactive immune system, and SARS-COV-2 are three prerequisites of the autoimmune disease triangle following COVID-19 infection - PubMed, accessed August 18, 2025, https://pubmed.ncbi.nlm.nih.gov/36182877/
Molecular Mimicry, Hyperactive Immune System, And SARS-COV-2 ..., accessed August 18, 2025, https://www.researchgate.net/publication/362843558_Molecular_Mimicry_Hyperactive_Immune_System_And_SARS-COV-2_Are_Three_Prerequisites_of_the_Autoimmune_Disease_Triangle_Following_COVID-19_Infection
Coronavirus associated molecular mimicry common to SARS-CoV-2 peptide - bioRxiv, accessed August 18, 2025, https://www.biorxiv.org/content/10.1101/2021.01.28.428642v1.full.pdf
Predicted structural mimicry of spike receptor-binding motifs from highly pathogenic human coronaviruses - PMC - PubMed Central, accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8249111/
Molecular Mimicry between SARS-CoV-2 and Human Endocrinocytes: A Prerequisite of Post-COVID-19 Endocrine Autoimmunity? - MDPI, accessed August 18, 2025, https://www.mdpi.com/1873-149X/29/3/39
Convergent Evolution Dynamics of SARS-CoV-2 and HIV Surface ..., accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9915253/
HIV-1 and SARS-CoV-2: Patterns in the evolution of two pandemic pathogens - PMC, accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8173590/
On Similarities and Differences of HIV and SARS Cov-2; Open Questions Relevant for COVID-19 Disease - ResearchGate, accessed August 18, 2025, https://www.researchgate.net/publication/356165334_On_Similarities_and_Differences_of_HIV_and_SARS_Cov-2_Open_Questions_Relevant_for_COVID-19_Disease
Uncanny similarity of unique inserts in the 2019-nCoV ... - bioRxiv, accessed August 18, 2025, https://www.biorxiv.org/content/10.1101/2020.01.30.927871v1.full.pdf
Uncanny similarity of unique inserts in the 2019-nCoV spike protein to HIV-1 gp120 and Gag, accessed August 18, 2025, https://www.biorxiv.org/content/10.1101/2020.01.30.927871v1
A nonself sugar mimic of the HIV glycan shield shows enhanced antigenicity - PNAS, accessed August 18, 2025, https://www.pnas.org/doi/10.1073/pnas.1002717107
Unraveling the SARS-CoV-2 spike protein long-term effect on neuro-PASC - Frontiers, accessed August 18, 2025, https://www.frontiersin.org/journals/cellular-neuroscience/articles/10.3389/fncel.2024.1481963/full
Dementia: What It Is, Causes, Symptoms, Treatment & Types - Cleveland Clinic, accessed August 18, 2025, https://my.clevelandclinic.org/health/diseases/9170-dementia
Cognitive deficits in PWH with post-acute sequelae of SARS-CoV-2 infection - SD CFAR, accessed August 18, 2025, https://cfar.ucsd.edu/media/filer_public/7d/d5/7dd557f1-2829-4519-a292-0a12876d052c/spies-cherner-protocol_full_20220203.pdf
Neuropsychiatric Symptoms of COVID-19 Explained by SARS-CoV-2 Proteins' Mimicry of Human Protein Interactions - Frontiers, accessed August 18, 2025, https://www.frontiersin.org/journals/human-neuroscience/articles/10.3389/fnhum.2021.656313/full
Protein Accumulation Linked to Neurological Symptoms of Long COVID, accessed August 18, 2025, https://www.technologynetworks.com/immunology/news/protein-accumulation-linked-to-neurological-symptoms-of-long-covid-393854
SARS-CoV-2 Spike Protein Accumulation in the Skull-Meninges-Brain Axis: Potential Implications for Long-Term Neurological Complications in post-COVID-19 | bioRxiv, accessed August 18, 2025, https://www.biorxiv.org/content/10.1101/2023.04.04.535604v1
SARS-CoV-2 spike protein found lingering in brain regions - News-Medical, accessed August 18, 2025, https://www.news-medical.net/news/20241203/SARS-CoV-2-spike-protein-found-lingering-in-brain-regions.aspx
SARS-CoV-2 Spike protein alters microglial purinergic signaling - PMC - PubMed Central, accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10126242/
Role of SARS-CoV-2 Spike-Protein-Induced Activation of Microglia ..., accessed August 18, 2025, https://www.mdpi.com/2073-4409/12/5/688
Microglial Implications in SARS-CoV-2 Infection and COVID-19: Lessons From Viral RNA Neurotropism and Possible Relevance to Parkinson's Disease - Frontiers, accessed August 18, 2025, https://www.frontiersin.org/journals/cellular-neuroscience/articles/10.3389/fncel.2021.670298/full
Elucidating the Neuropathologic Mechanisms of SARS-CoV-2 Infection - PubMed Central, accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8072392/
Pathological Features and Neuroinflammatory Mechanisms of SARS-CoV-2 in the Brain and Potential Therapeutic Approaches - PubMed Central, accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9313047/
HIV gp120-induced neuroinflammation potentiates NMDA receptors ..., accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6379117/
Effects of HIV gp120 on neuroinflammation in immunodeficient vs. immunocompetent states, accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7785647/
Neuroinflammation, Blood–Brain Barrier, and HIV Reservoirs in the CNS: An In-Depth Exploration of Latency Mechanisms and Emerging Therapeutic Strategies - MDPI, accessed August 18, 2025, https://www.mdpi.com/1999-4915/17/4/572
Early Insights into COVID-19 in Persons Living with HIV and Cardiovascular Manifestations, accessed August 18, 2025, https://www.scientificarchives.com/article/early-insights-into-covid-19-in-persons-living-with-hiv-and-cardiovascular-manifestations
Macrovascular Function in People with HIV After Recent SARS-CoV ..., accessed August 18, 2025, https://www.mdpi.com/2813-2475/4/1/4
Coagulation Disorders and Thrombosis in COVID-19 Patients and a ..., accessed August 18, 2025, https://www.aginganddisease.org/EN/10.14336/AD.2021.0704
Coagulation Disorders and Thrombosis in COVID-19 Patients and a Possible Mechanism Involving Endothelial Cells: A Review - ResearchGate, accessed August 18, 2025, https://www.researchgate.net/publication/358346643_Coagulation_Disorders_and_Thrombosis_in_COVID-19_Patients_and_a_Possible_Mechanism_Involving_Endothelial_Cells_A_Review
Challenging the notion of endothelial infection by SARS-CoV-2: insights from the current scientific evidence - Frontiers, accessed August 18, 2025, https://www.frontiersin.org/journals/immunology/articles/10.3389/fimmu.2025.1443932/full
Endothelial dysfunction in long-COVID: New insights from the nationwide multicenter LINCOLN Study | Request PDF - ResearchGate, accessed August 18, 2025, https://www.researchgate.net/publication/364181643_Endothelial_Dysfunction_in_Long-COVID_New_Insights_from_the_Nationwide_Multicenter_LINCOLN_Study
Myocarditis With COVID-19 mRNA Vaccines | Circulation, accessed August 18, 2025, https://www.ahajournals.org/doi/10.1161/CIRCULATIONAHA.121.056135
Molecular Mimicry of the Viral Spike in the SARS-CoV-2 Vaccine Possibly Triggers Transient Dysregulation of ACE2, Leading to Vascular and Coagulation Dysfunction Similar to SARS-CoV-2 Infection - PubMed Central, accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10222462/
The spike protein downregulates ACE2 that in turn increases Ang II and... - ResearchGate, accessed August 18, 2025, https://www.researchgate.net/figure/The-spike-protein-downregulates-ACE2-that-in-turn-increases-Ang-II-and-decreases-Ang_fig1_349148910
HIV Proteins and Endothelial Dysfunction: Implications in Cardiovascular Disease - Frontiers, accessed August 18, 2025, https://www.frontiersin.org/journals/cardiovascular-medicine/articles/10.3389/fcvm.2018.00185/full
The Relationship Between HIV Infection and Cardiovascular Disease - PubMed Central, accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC2780822/
Vascular endothelial growth factor A: friend or foe in the pathogenesis of HIV and SARS-CoV-2 infections? - Frontiers, accessed August 18, 2025, https://www.frontiersin.org/journals/cellular-and-infection-microbiology/articles/10.3389/fcimb.2024.1458195/full
Vascular endothelial growth factor A: friend or foe in the pathogenesis of HIV and SARS-CoV-2 infections? - ResearchGate, accessed August 18, 2025, https://www.researchgate.net/publication/388909346_Vascular_endothelial_growth_factor_A_friend_or_foe_in_the_pathogenesis_of_HIV_and_SARS-CoV-2_infections
Health & Medicine News - ScienceDaily, accessed August 18, 2025, https://www.sciencedaily.com/news/health_medicine/
(PDF) Plasmatic HIV-1 soluble gp120 is associated with immune dysfunction and inflammation in ART-treated individuals with undetectable viremia - ResearchGate, accessed August 18, 2025, https://www.researchgate.net/publication/373271562_Plasmatic_HIV-1_soluble_gp120_is_associated_with_immune_dysfunction_and_inflammation_in_ART-treated_individuals_with_undetectable_viremia
Plasma Human Immunodeficiency Virus 1 Soluble Glycoprotein 120 Association With Correlates of Immune Dysfunction and Inflammation in Antiretroviral Therapy–Treated Individuals With Undetectable Viremia | The Journal of Infectious Diseases | Oxford Academic, accessed August 18, 2025, https://academic.oup.com/jid/article/229/3/763/7453367
In Silico and In Vitro Evaluation of the Molecular Mimicry of the SARS-CoV-2 Spike Protein by Common Short Constituent Sequences (cSCSs) in the Human Proteome: Toward Safer Epitope Design for Vaccine Development - MDPI, accessed August 18, 2025, https://www.mdpi.com/2076-393X/12/5/539
SARS-CoV-2-Induced Immunosuppression: A Molecular Mimicry Syndrome - PMC, accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9282940/
Molecular Mimicry of SARS-CoV-2 Spike Protein in the Nervous System: A Bioinformatics Approach - PubMed Central, accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9605789/
Human Immunodeficiency Virus Proteins Mimic Human T Cell Receptors Inducing Cross-Reactive Antibodies - PMC, accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5666773/
Autoimmune diseases and HIV infection: A cross-sectional study - PMC - PubMed Central, accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5287948/
Role of HIV-1 Tat Protein Interactions with Host Receptors in HIV Infection and Pathogenesis, accessed August 18, 2025, https://www.mdpi.com/1422-0067/25/3/1704
HIV-1 Tat: Role in Bystander Toxicity - Frontiers, accessed August 18, 2025, https://www.frontiersin.org/journals/cellular-and-infection-microbiology/articles/10.3389/fcimb.2020.00061/full
HIV-Associated Neurocognitive Disorder (HAND) and Alzheimer's ..., accessed August 18, 2025, https://www.mdpi.com/1422-0067/25/20/11170
Mechanisms of HIV-1 Tat Neurotoxicity via CDK5 Translocation and ..., accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4455959/
Neurotoxicity of HIV-1 Tat protein: Involvement of D1 dopamine receptor - PMC, accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC2957183/
HIV-1 Tat: Molecular Switch in Viral Persistence and Emerging Technologies for Functional Cure - MDPI, accessed August 18, 2025, https://www.mdpi.com/1422-0067/26/13/6311
Evidence for neurotoxic activity of Tat from human immunodeficiency virus type 1, accessed August 18, 2025, https://www.researchgate.net/publication/21256963_Evidence_for_neurotoxic_activity_of_Tat_from_human_immunodeficiency_virus_type_1
HIV-1 Tat and Viral Latency: What We Can Learn from Naturally Occurring Sequence Variations - Frontiers, accessed August 18, 2025, https://www.frontiersin.org/journals/microbiology/articles/10.3389/fmicb.2017.00080/full
Human Immunodeficiency Virus Type-1 Protein Tat Induces Tumor Necrosis Factor-α-Mediated Neurotoxicity - PMC - PubMed Central, accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC2080622/
Neuroinflammation and COVID-19 - PMC - PubMed Central, accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9239981/
Structure, function, and inhibitor targeting of HIV-1 Nef-effector kinase complexes - PMC, accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7606690/
HIV-1 Nef and T-cell activation: a history of contradictions - PMC, accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3810967/
How HIV Nef Proteins Hijack Membrane Traffic To Promote Infection - PMC, accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6880166/
Proposed model of the involvement of HIV-1 Nef and TGFβ-1 in... - ResearchGate, accessed August 18, 2025, https://www.researchgate.net/figure/Proposed-model-of-the-involvement-of-HIV-1-Nef-and-TGFb-1-in-neuroinflammation-and_fig5_337903215
Viral Mimicry of Interleukin-17A by SARS-CoV-2 ORF8 | mBio - ASM Journals, accessed August 18, 2025, https://journals.asm.org/doi/10.1128/mbio.00402-22
Predicting human and viral protein variants affecting COVID-19 susceptibility and repurposing therapeutics | bioRxiv, accessed August 18, 2025, https://www.biorxiv.org/content/10.1101/2023.11.07.566012v1.full-text
Variation in HIV-1 Tat and Vpr protein amino acid sequences and its association with vascular health measures in a South African cohort: an exploratory study, accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC12323018/
HIV-1 Vpr suppresses immune activation and apoptosis through regulation of nuclear factor kappa B - PubMed, accessed August 18, 2025, https://pubmed.ncbi.nlm.nih.gov/9334723/
HIV viral protein R causes atrial cardiomyocyte mitosis, mesenchymal tumor, dysrhythmia, and heart failure - PubMed, accessed August 18, 2025, https://pubmed.ncbi.nlm.nih.gov/15608661/
Role of HIV-encoded proteins in cardiovascular disease - PMC, accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC12309391/
Retrotransposition and senescence in mouse heart tissue by viral protein R of human immunodeficiency virus-1 - PubMed, accessed August 18, 2025, https://pubmed.ncbi.nlm.nih.gov/32243892/
COVID-19-Induced Myocarditis: Pathophysiological Roles of ACE2 and Toll-like Receptors, accessed August 18, 2025, https://www.mdpi.com/1422-0067/24/6/5374
CCL2-mediated endothelial injury drives cardiac dysfunction in long COVID - PubMed, accessed August 18, 2025, https://pubmed.ncbi.nlm.nih.gov/39402206/
Species-Specific Activity of SIV Nef and HIV-1 Vpu in Overcoming Restriction by Tetherin/BST2 | PLOS Pathogens, accessed August 18, 2025, https://journals.plos.org/plospathogens/article?id=10.1371/journal.ppat.1000429
HIV-1 Vpu – An ion channel in search of a job - PMC - PubMed Central, accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3859730/
Role of host immune responses in sequence variability of HIV-1 Vpu, accessed August 18, 2025, https://www.wjgnet.com/2219-2824/full/v4/i2/107.htm
Role of Viral Protein U (Vpu) in HIV-1 Infection and Pathogenesis ..., accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8402909/
Differential dysregulation of β-TrCP1 and -2 by HIV-1 Vpu leads to ..., accessed August 18, 2025, https://pubmed.ncbi.nlm.nih.gov/37341489/
HIV Vpu Interferes with NF-κB Activity but Not with Interferon Regulatory Factor 3, accessed August 18, 2025, https://journals.asm.org/doi/10.1128/jvi.01596-15
Targeting inflammasomes as a therapeutic potential for HIV/AIDS - PMC - PubMed Central, accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC12006635/
(PDF) Inflammasomes in Human Immunodeficiency Virus Type 1 Infection - ResearchGate, accessed August 18, 2025, https://www.researchgate.net/publication/368309518_Inflammasomes_in_Human_Immunodeficiency_Virus_Type_1_Infection
Tat-Vpu Peptide Inducing a Shock Effect on Human Immunodeficiency Virus Type-1 Infected Primary Macrophage Cells and Understanding a Novel NLRP3 Inflammasome Activation Induced by GU-Rich Human Immunodeficiency Virus Type-1 ssRna - ProQuest, accessed August 18, 2025, https://search.proquest.com/openview/582a10cf23d918108a74fb4bb0e8a635/1?pq-origsite=gscholar&cbl=18750&diss=y
Long-Term SARS-CoV-2 Infection in a Person with HIV Offers New Clues to Viral Evolution, accessed August 18, 2025, https://globalbiodefense.com/2025/08/02/long-term-sars-cov-2-infection-in-a-person-with-hiv-offers-new-clues-to-viral-evolution/
New Evidence Supports Autoimmunity as One of Long COVID's Underlying Drivers, accessed August 18, 2025, https://medicine.yale.edu/news-article/new-evidence-supports-autoimmunity-as-one-of-long-covids-underlying-drivers/
Autoantibodies in COVID-19 survivors with post-COVID symptoms: a systematic review, accessed August 18, 2025, https://www.frontiersin.org/journals/immunology/articles/10.3389/fimmu.2024.1428645/full
Is Long Covid a New Autoimmune Disease?, accessed August 18, 2025, https://www.autoimmuneinstitute.org/articles/is-long-covid-a-new-autoimmune-disease/
A clinical trial of repurposed HIV antivirals in LongCOVID - PolyBio ..., accessed August 18, 2025, https://polybio.org/projects/a-clinical-trial-of-repurposed-hiv-antivirals-in-longcovid/
Investigating the feasibility of repurposing HIV antivirals in Adults with Long Covid | Mount Sinai - New York, accessed August 18, 2025, https://www.mountsinai.org/clinical-trials/investigating-the-feasibility-of-repurposing-hiv-antivirals-in-adults-with-long-covid
Study Details | Antiviral Clinical Trial for Long Covid-19 - ClinicalTrials.gov, accessed August 18, 2025, https://clinicaltrials.gov/study/NCT06511063?term=putrino&rank=1
COVID-19 drug repurposing research - Wikipedia, accessed August 18, 2025, https://en.wikipedia.org/wiki/COVID-19_drug_repurposing_research
Treatment on Trial to Reverse Long COVID Effects - Discoveries in Medicine, accessed August 18, 2025, https://discoveries.vanderbilthealth.com/2025/01/treatment-on-trial-to-reverse-long-covid-effects/
Harnessing immunity: Immunomodulatory therapies in COVID-19 - PMC - PubMed Central, accessed August 18, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11229839/
Immune Immunomodulation in Coronavirus Disease 2019 (COVID-19): Strategic Considerations for Personalized Therapeutic Intervention - Oxford Academic, accessed August 18, 2025, https://academic.oup.com/cid/article/74/1/144/5865455
Immunomodulatory therapies for COVID-19 - Frontiers, accessed August 18, 2025, https://www.frontiersin.org/journals/medicine/articles/10.3389/fmed.2022.921452/full