Next Article in Journal
Syndromic Testing in the Pandemic Era and Beyond: Rapid Detection for Respiratory Infections in Istanbul
Previous Article in Journal
Seroconversion Is Misleading as a Test for HSV-2 Infection in Prophylactic Genital Herpes Vaccine Trials: Results of Vaccine Studies in Guinea Pigs
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Finetuning Type I Interferon Signaling to Enhance T Cell Immunity in HIV Infection

Division of Hematology and Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
*
Author to whom correspondence should be addressed.
Viruses 2025, 17(6), 774; https://doi.org/10.3390/v17060774
Submission received: 18 April 2025 / Revised: 20 May 2025 / Accepted: 26 May 2025 / Published: 29 May 2025
(This article belongs to the Special Issue Interferon Signaling in Viral Pathogenesis)

Simple Summary

Type I interferons (IFN-Is) have a complex role in HIV-1 infection, offering early protection but contributing to harm over time. In the acute phase, IFN-Is help control the virus, activate the innate immune system, and reduce the formation of long-term viral reservoirs. However, during chronic HIV-1 infection, continued IFN-I signaling can become harmful. It leads to ongoing inflammation, T cell exhaustion, and tissue damage. Prolonged IFN-I exposure also increases immune suppression by upregulating inhibitory receptors on T cells and disrupting their metabolism. New therapies are being developed to balance IFN-I’s benefits and drawbacks. These include blocking IFN-I receptors (IFNARs), enhancing autophagy, inhibiting JAK-STAT signaling, and combining immune checkpoint therapies. Such strategies aim to reduce immune dysfunction and improve T cell responses. Overall, understanding and adjusting IFN-I signaling could help manage chronic HIV-1 more effectively, minimizing harm while preserving its antiviral benefits.

Abstract

Type I interferons (IFN-Is) play a dual role in the immune response to HIV-1, providing early antiviral defense while driving immune dysfunction in the chronic phase. During acute infection, robust IFN signaling is critical in controlling viral replication, activating innate immunity, and limiting reservoir establishment. However, sustained IFN-I activation during chronic infection fuels systemic inflammation, immune exhaustion, and fibrosis, particularly in lymphoid tissues such as gut-associated lymphoid tissue (GALT). Prolonged IFN-I exposure upregulates inhibitory receptors on T cells, impairs metabolic fitness, and fosters an immunosuppressive cytokine milieu that weakens overall immune responses. In contrast to natural SIV (Simian immunodeficiency virus) hosts, IFN-I responses are tightly regulated to prevent chronic immune activation and tissue damage. However, humans and non-natural hosts experience persistent Interferon Stimulated Gene (ISG) expression and IFN-I driven inflammation. Emerging therapeutic strategies seek to harness the antiviral benefits of IFN-I while mitigating its pathogenic effects. Approaches such as the IFNAR blockade, autophagy induction, JAK-STAT inhibition, and combined immune inhibitory blockade therapy show promise in restoring immune balance and enhancing T cell function. This review examines the mechanisms of IFN-I dysregulation in chronic HIV-1 infection and highlights novel interventions aimed at finetuning IFN-I signaling for therapeutic benefit.

1. Introduction

Human immunodeficiency virus (HIV) remains a major global health challenge, affecting over 38 million individuals worldwide [1,2]. Despite the substantial success of antiretroviral therapy (ART) in controlling viral replication and reducing mortality, immune dysfunction remains a major challenge in people living with HIV (PLWH) [3,4,5]. One of the critical drivers of this immune dysregulation is the dysregulated activation of type I interferon (IFN-I) signaling [6,7,8].
Type I IFNs, including IFN-α and IFN-β, are pivotal to the host’s antiviral defense, orchestrating early immune responses by stimulating the expression of interferon-stimulated genes (ISGs) [9,10,11,12]. The induction of type I IFN signaling occurs upon the recognition of specific ligands, such as viral nucleic acid or viral proteins by pattern recognition receptors (PRRs) such as Toll-like receptors (TLRs), RIG-I receptors (RLRs), NOD-like receptors (NLRs), etc., in response to viral infection [13]. The recognition of PRRs activates signaling pathways that lead to the production of IFN-I, which diffuses into the extracellular space. IFN-I binds to the IFN receptor (IFNAR) on the surface of nearby cells, including immune cells and epithelial cells [11]. The binding of IFN-I to IFNAR activates the JAK-STAT signaling pathway [11]. Upon binding, IFNAR1-associated protein tyrosine kinase 2 (Tyk2) and IFNAR2-associated protein tyrosine kinase 1 (JAK1) initiate phosphorylation events that activate a range of signal transducer and activator of transcription (STAT) proteins [11]. These activated STAT proteins then dimerize and translocate to the nucleus [11]. In the nucleus, phosphorylated STAT proteins bind to interferon-stimulated response elements (ISREs) and activate the transcription of ISGs [10]. ISGs encode a diverse set of proteins with antiviral functions, including the inhibition of viral entry, reverse transcription, and release; the degradation of viral RNA; and the modulation of host immune responses [9,12]. For example, IFITM (interferon-induced transmembrane) proteins block viral entry [14], ISG15 functions as a ubiquitin-like modifier that is conjugated to viral and host proteins, affecting their stability or localization [15], and OAS (2′-5′-oligoadenylate synthetase) activates RNase L, leading to the degradation of viral RNA [16]. Mx proteins are GTPases that trap viral nucleocapsids and prevent their nuclear import or replication [17]. These genes are essential for controlling viral replication during the acute phase of infection [18,19,20,21,22].
Despite its critical role in antiviral response, chronic type I IFN signaling is involved in driving chronic inflammation and immune exhaustion [23]. A defining feature of chronic HIV infection is the persistence of type I IFN responses, characterized by sustained ISG expression and prolonged immune activation [24,25,26,27,28]. In humans, the persistent activation of type I IFN pathways during chronic HIV infection leads to the upregulation of inhibitory receptors such as PD-1, TIM-3, and LAG-3 on T cells [29,30]. This contributes to immune exhaustion, characterized by diminished T cell proliferation, impaired cytokine production, and reduced cytotoxic activity [31,32,33,34]. Chronic IFN-I signaling also disrupts immune homeostasis by promoting systemic inflammation, tissue fibrosis, and metabolic dysregulation in key lymphoid compartments such as gut-associated lymphoid tissue (GALT) [33,35,36,37]. These pathological changes compromise immune restoration even in individuals receiving long-term ART. This review explores the multifaceted role of type I IFN signaling in HIV infection, with a particular focus on its dualistic impact on T cell immunity. While early IFN-I responses are critical for limiting viral replication and activating innate immunity, their prolonged activation contributes to immune suppression and disease progression. Emerging therapeutic strategies aimed at modulating type I IFN responses offer potential avenues to restore immune function, enhance T cell efficacy, and improve clinical outcomes in individuals living with HIV.

2. Early Protective Roles of Type I IFNs During Acute HIV Infection

Type I IFNs, including IFN-α and IFN-β, are critical cytokines that mediate the early host immune response to HIV infection [11,38]. During the acute phase of HIV infection, type I IFNs are rapidly produced by infected cells and plasmacytoid dendritic cells (pDCs) upon sensing viral RNA through PRRs, including endosomal TLR7 and such as cytosolic sensors (e.g., RLRs and cGAS-STING) [39,40]. This production of type I IFNs initiates a cascade of signaling pathways through the JAK-STAT pathway, leading to the induction of hundreds of ISGs [10,41]. These ISGs are responsible for targeting and inhibiting various stages of the HIV lifecycle, contributing to the early containment of the virus [42,43]. One well-studied ISG is tetherin (BST2), a protein that restricts viral particle release by anchoring budding virions to the host cell membrane, preventing their dissemination [42,44,45]. Research has shown that tetherin expression is upregulated in response to IFN-I signaling, effectively reducing the spread of HIV during early infection stages [44]. Similarly, APOBEC3G, an ISG and cytidine deaminase, induces hypermutation in the viral genome during reverse transcription in the absence of HIV viral protein Vif, rendering the virus replication incompetent [46,47,48]. Another critical ISG, SAMHD1, restricts viral replication by depleting the pool of deoxynucleotide triphosphates (dNTPs) required for reverse transcription. Studies have demonstrated that SAMHD1-mediated dNTP depletion, induced by type I IFNs, significantly limits HIV replication in macrophages and dendritic cells [49,50].
Type I IFNs are pivotal not only in initiating antiviral defenses but also in amplifying and coordinating the innate immune response. For example, they enhance the cytotoxic activity of natural killer (NK) cells, enabling them to target and kill infected cells more effectively [51,52]. Type I IFNs upregulate the expression of ligands for activating NK cell receptors, such as NKG2D, thereby increasing their susceptibility to NK-mediated killing during chronic viral infection [53,54]. Furthermore, IFN-Is stimulate pDCs, enhancing their antigen-presenting capacity and facilitating the activation of adaptive immune responses [55,56,57]. IFN-α has been shown to improve DC maturation and upregulate the expression of major histocompatibility complex (MHC) molecules, which are essential for T cell activation [58,59,60].
Robust type I IFN responses play a pivotal role in controlling acute HIV infection. Studies have shown that, during primary HIV infection, a high plasma viral load triggers a rapid but transient increase in multiple cytokines and chemokines, particularly type I IFNs [61]. This early IFN-I surge enhances antiviral immunity by stimulating the immune system and inducing antiviral factors, ultimately leading to lower viral loads. Individuals with a higher ISG expression in the early stages of HIV infection tend to exhibit lower viral loads and slower disease progression [62]. A clinical study by Stacey et al. on acute HIV patients monitored plasma viral load prior to infection and found that IFN-α and its mediators rise weeks before peak viremia, suggesting a predictive role in early immune responses [61]. Additionally, in asymptomatic HIV-infected individuals, IFN-α treatment has been shown to reduce viral isolation frequency and slow disease progression [63].
Collectively, these findings underscore the critical protective role of type I IFNs in acute HIV infection. However, the relationship between IFN levels and HIV disease progression is complex, influenced by factors such as specific IFN genes activated, downstream gene expression patterns, and the infection stage [31,64]. These findings highlight the critical role of type I IFNs in the host defense against HIV and their potential as therapeutic targets. Modulating IFN responses to enhance their protective effects while minimizing their detrimental consequences during chronic infection is an area of active research.

3. Chronic Dysregulation of Type I IFN Signaling

In contrast to its beneficial role in early infection, persistent IFN signaling during chronic HIV infection drives immune activation and dysfunction, leading to T cell and NK cell exhaustion and systemic inflammation. This chronic activation is fueled by low-level viral replication and microbial translocation from the gut, which continuously stimulates IFN-I production [65]. Non-human primate studies provided the first evidence linking excessive type I IFN responses to chronic immune activation. Natural hosts of Simian immunodeficiency virus (SIV), such as African green monkeys and sooty mangabeys, exhibit a controlled immune response where ISG expression spikes during acute infection but is quickly downregulated in the chronic phase, allowing them to avoid long-term immune activation [66,67,68]. In contrast, non-natural hosts like macaques experience prolonged ISG activation, which drives chronic inflammation and progression to AIDS [69,70,71,72]. These early findings demonstrated how an unregulated type I IFN response can transform an initially protective antiviral mechanism into a driver of immune dysfunction.
Similar patterns have been observed in humans. As HIV-1 infection progresses from the acute to the chronic phase, type I IFN signaling shifts from a protective antiviral response to a driver of immune pathology. Persistently elevated levels of ISGs are closely associated with CD4 T cell depletion and altered T cell dynamics [73,74]. For example, high levels of ISG15, an IFN-stimulated gene, were found to correlate with high viral loads and low CD4+ cell counts in patients with HIV [75]. The prolonged type I IFN responses in conjunction with sustained alterations in CD4+ and CD8+ T cell signaling pathways [76] exacerbate immune exhaustion and promote chronic inflammation—hallmarks of chronic HIV-1 infection [30,77,78,79].
Interestingly, several studies have reported that women exhibit a stronger IFN-I response during the acute phase of HIV infection, resulting in a lower viral reservoir compared to men [80]. One key reason for this difference is attributed to the increased capacity of pDCs from females to produce IFN-α upon TLR7 stimulation. During HIV-1 infection, TLR7 activation in pDCs drives robust IFN-I production. Notably, the TLR7 gene escapes X-chromosome inactivation in the immune cells of females, leading to higher TLR7 expression and stronger IFN-1 signaling compared to males [81,82]. Additionally, ISGs such as MX1, IFIT1, IFIT3, ISG15, OAS2, and IRF7 are more highly expressed in T cells and dendritic cells of HIV-infected women [83]. Despite this early advantage, women progress to AIDS faster than men at equivalent viral loads [84,85]. These findings underscore the dual role of IFN-I signaling in HIV infection—beneficial in early control but could be detrimental when chronically activated.
Persistent type I IFN signaling during chronic HIV infection induces the sustained expression of inhibitory receptors such as PD-1, TIM-3, and LAG-3 on CD4+ and CD8+ T cells [29,86,87], which impair T cell activation, proliferation, and effector functions, ultimately leading to immune exhaustion [26,88,89]. Prolonged IFN signaling also shifts cellular metabolism toward oxidative phosphorylation, reducing the glycolytic activity required for effective T cell proliferation and effector function [90,91]. This metabolic dysregulation contributes to the reduced functionality and survival of HIV-specific T cells, exacerbating immune exhaustion [90,91]. Persistent type I IFN also promotes the release of inflammatory cytokines, such as IL-10 and TGF-β, which are known to suppress T cell activity and reinforce an immunosuppressive environment [23,92,93,94]. Moreover, IFN-induced pro-apoptotic signals promote the premature death of activated T cells, diminishing the pool of functional HIV-specific T cells [95,96]. Prolonged type I IFN signaling creates a feedback loop of immune suppression and inflammation in chronic HIV infection, leading to T cell exhaustion, metabolic dysregulation, and cytokine-mediated suppression, all of which impair the immune system’s ability to control HIV replication and viral reservoirs [90].
Building on these systemic effects, recent studies have shown that type I IFN responses display tissue-specific dynamics, which also play a critical role in disease pathogenesis during chronic HIV infection. IFN-I, particularly IFNα, exhibit localized expression patterns during HIV-1 pathogenesis, with significantly higher expression in lymph node tissues compared to peripheral blood during chronic untreated infection. Despite ART, type I IFN signaling persists in gut tissues, likely driven by the early migration of pDCs—a key source of IFN-I—to mucosal and lymphoid sites during acute infection [97]. The sustained IFN-I activity fosters a chronic inflammatory environment that contributes to tissue fibrosis in lymphoid organs, including lymph nodes and GALT [98,99,100,101]. Fibrosis disrupts lymphoid architecture and impairs cell-to-cell interactions necessary for effective immune responses, leading to further CD4⁺ T cell depletion and immune dysfunction [98,99,102]. Persistent IFN-I signaling in GALT may exacerbate gut integrity and promote microbial translocation and further amplify systemic inflammation [103]. This chronic immune activation, driven in part by IFN-I, not only sustains viral reservoirs but also contributes to non-AIDS-related comorbidities, such as cardiovascular disease and metabolic disorders, even in individuals on suppressive ART [79,104]. Understanding these interactions between IFN-I, tissue microenvironments, and microbial factors could pave the way for novel interventions aimed at reducing inflammation and improving long-term outcomes in PLWH.
Primate studies further underscore the double-edged sword nature of type I IFN responses. Blocking IFNAR during acute SIV infection in rhesus macaques worsened outcomes by increasing reservoir size, decreasing antiviral gene expression, and accelerating disease progression, highlighting the importance of early IFN-I responses [69]. Conversely, prolonged IFNα administration led to desensitization, reduced ISG expression, and accelerated CD4+ T cell depletion, illustrating the harmful effects of sustained signaling [69]. However, blocking IFNAR in combination with ART during the chronic phase enhanced viral suppression, reduced reservoirs, and restored CD8+ T cell functions [105]. In sum, these findings suggested the complex role of type I IFNs in HIV-1 infection, highlighting that finetuning IFN-I responses may offer therapeutic benefits by balancing early immune activation with the prevention of long-term immune dysfunction.

4. Mechanisms of Immune Response Impairment by Type I IFNs

Type I IFNs significantly influence immune cell metabolism, particularly by inducing metabolic reprogramming to support immune activation. IFN-I helps promote glycolysis in immune cells, including macrophages and dendritic cells, enabling rapid and early innate immune responses to meet the increased energy demands during immune activation [91,106,107]. This metabolic shift allows immune cells to generate ATP quickly, ensuring that they have the energy necessary for their functions. For instance, Pantel et al. demonstrated that the direct activation of dendritic cells by IFN-I leads to a metabolic shift towards glycolysis, which is essential for their maturation and function [106]. Hedl et al. have shown that IRF5 can lead to increased glycolysis (a metabolic pathway) and promote the polarization of human macrophages towards an inflammatory M1 phenotype [107]. Additionally, IFN-I can modulate glucose metabolism through PI3K/Akt signaling, which has been shown to be essential for antiviral responses, as observed in the regulation of glucose metabolism during Coxsackievirus B3 infection [108].
However, excessive reliance on glycolysis can compromise mitochondrial function, leading to reduced oxidative phosphorylation and impaired energy production [109,110]. Lewis et al. showed that IFN-I inhibits mitochondrial activity, which may impair cellular energy production, and this has been observed in CD4+ T cells treated with IFN-β, which correlates with impaired mitochondrial function in diseases like multiple sclerosis [111,112]. Olson et al. have also shown that the induction of IFN-β correlates with impaired mitochondrial function, which potentially disrupts this balance between glycolysis and oxidative phosphorylation, critical for maintaining efficient cellular energy metabolism [113]. In juvenile systemic lupus erythematosus (SLE), a chronic autoimmune disease, type I IFN signaling has been associated with mitochondrial dysfunction in cytotoxic CD8+ T cells [114]. CD8+ T cells from IFN-high SLE patients exhibit enlarged mitochondria and reduced respiratory capacity, while exposure to IFNα increases NAD+ consumption. Prolonged IFNα exposure depletes NAD+, a key molecule for mitochondrial function, further impairing the energy production capacity of CD8+ T cells [114]. Additionally, type I IFNs elevate the production of reactive oxygen species (ROS) in T cells [114] and hematopoietic cells [115], further compromising mitochondrial integrity and function. Therefore, those studies highlighted the detrimental effects of sustained IFN-I activity on mitochondrial health.
Type I IFNs reshape the cytokine environment by promoting the production of pro-inflammatory cytokines such as IL-6, TNF-α, and IL-12 [61,116]. These cytokines indirectly support early antiviral defenses by enhancing Th1 polarization and increasing IFN-γ production through STAT4 activation [117,118,119]. Additionally, they promote the recruitment of cytotoxic CD8+ T cells via the TNF-α-induced upregulation of adhesion molecules like ICAM-1, facilitating an early immune response against HIV infection [120]. Furthermore, IFN-I provides a third signal to CD8+ T cells through a STAT4-dependent pathway, promoting their survival, cytolytic function, and IFN-γ production [121]. However, sustained IFN-I signaling in chronic infection triggers the excessive production of pro-inflammatory mediators (e.g., IL-6, CXCL10) while creating an immunosuppressive environment that hinders effective T cell responses. One of the primary ways is through the induction of anti-inflammatory cytokine IL-10 [122]. Interleukin-10 (IL-10) plays a crucial role in suppressing T cell responses and inducing immune tolerance. IL-10 directly inhibits T cell proliferation and cytokine production [123]. IL-10 suppresses T cell responses through the downregulation of APC co-stimulatory molecules (CD80/CD86) and the inhibition of DC-derived IL-12 (via SOCS3-mediated NF-κB suppression) [124]. In addition, TGF-β utilizes various mechanisms to inhibit the proliferation and reactivity of T lymphocytes by the suppression of IL-2 production, the downregulation of c-myc, and the suppression of T cell responses by regulating Foxp3+ Tregs [125,126,127,128]. Together, these cytokine-driven changes create a feedback loop of immune suppression that limits T cells’ ability to mount robust responses against viral replication. Altogether, this explains why the altered cytokine landscape is a key contributor to the inability to control HIV replication and clear latent reservoirs [129,130].
Beyond altering soluble mediators, type I IFNs indirectly impair immune responses by modulating antigen-presenting cells (APCs) themselves. Chronic IFN-I exposure skews monocytes and dendritic cells toward a dysregulated or suppressive phenotype [131]. In persistent viral infection models, IFN-I signaling drives the differentiation of “immunoregulatory” DCs that produce IL-10 and express inhibitory ligands such as PD-L1 while simultaneously blunting the generation of conventional DCs with T cell-stimulatory capacity [132]. These IFN-I conditioned APCs can attenuate T cell activation through multiple pathways. Notably, the upregulated PD-L1 on DCs and macrophages engages PD-1 on T cells, reinforcing T cell exhaustion and functional inactivation [132]. IFN-I signaling in DCs also induces immunosuppressive enzymes like indoleamine 2,3-dioxygenase (IDO), which catabolizes tryptophan and thereby stifles T cell proliferation as a negative feedback mechanism [133].
Concomitantly, persistent IFN-I-driven inflammation alters the architecture of lymphoid tissues, further impairing immune competence. During chronic HIV/SIV infection, lymph nodes and gut-associated lymphoid tissue undergo structural alterations, including fibrosis and the disorganization of tissue architecture within the organs [100,134,135]. Prolonged IFN-I and inflammatory cytokines (e.g., TNF-α) stimulate fibroblastic reticular cells and stromal cells within lymphoid tissues to produce excess collagen and other extracellular matrix proteins [136], leading to fibrotic scarring of the T cell zones [137]. This fibrosis, which is largely absent in natural SIV hosts that tightly regulate type I IFN responses, correlates with the collapse of the follicular dendritic cell network in germinal centers and a loss of normal lymphoid tissue organization [100,138]. The outcome is a lymphoid infrastructure less capable of supporting effective immune surveillance. The disruption of stromal scaffolds and chemokine gradients further limits T cell positioning and survival, thereby exacerbating the immune dysfunction driven by chronic IFN-I signaling.
Type I IFNs, in cooperation with TNF, induce the tandem occupancy of regulatory elements at non-ISGs that encode inflammatory mediators by IRFs and NF-κB, a process associated with increased histone marks that facilitate transcription, leading to faster and higher ISG production in response to subsequent environmental rechallenges [139,140]. When T cells are continuously exposed to a chronic inflammatory environment, they undergo extensive epigenetic changes, including DNA methylation, histone acetylation, and chromatin remodeling, resulting in long-lasting alterations to their gene expression profiles and functional states [141]. Studies have shown that exhaustion genes, including PD-1, LAG-3, and TIM-3, are epigenetically modulated by type I IFNs, potentially leading to a sustained expression of these inhibitory receptors [29]. The expressions of IFNs and ISGs are quantitatively controlled in a cell lineage-specific manner by H3K9me2, a negative histone marker. Weak producers of IFN (basal level) have significantly lower levels of H3K9me2 at IFNs and ISG promoters [142], while higher levels of H3K9 methylation lead to a decrease in inflammatory chemokine production [143]. Additionally, stimulation with type I IFNs increases the amount of trimethylated histone H3 Lys 4 (H3K4me3), a histone mark that promotes transcription at the promoters of genes encoding inflammatory mediators [141]. These data show the potential of the epigenetic landscape induced by IFN-Is to act in contradictory ways, as one way can be protective, while another could exacerbate the inflammatory environment and cause immune cell dysfunction. These findings support further investigation into the epigenomic signatures and the relationships among IFN-I signaling, chromatin changes, and DNA methylation in the context of HIV infection.
The interplay of metabolic dysregulation, cytokine- and APC-driven suppression, epigenetic reprogramming, and lymphoid tissue remodeling creates a multifaceted impairment of T cell function under chronic Type I IFN exposure. Together, these mechanisms synergize to drive profound T cell exhaustion, characterized by reduced proliferative capacity, diminished cytokine production, and impaired cytotoxicity. Understanding the specific mechanisms by which type I IFNs impair T cell function provides critical insights into potential therapeutic approaches to fine-tune IFN-I signaling and restore immune competence.

5. Potential Therapeutic Strategies to Modulate Type I IFN Signaling in HIV Infection

The therapeutic modulation of IFN-I signaling primarily employs monoclonal antibodies targeting the IFNAR or its ligands and IFNAR antagonists specifically designed to block IFN-I binding, thereby suppressing the activation of downstream signaling cascades. This approach has shown significant promise in preclinical models of chronic viral infections [23,144,145]. In HIV infection, blocking IFNARs has been shown to restore the functionality of T cells, decrease inflammatory cytokine production, and enhance viral suppression. Studies in humanized mouse models of HIV infection demonstrated that IFNAR antagonists reduce immune exhaustion markers like PD-1 and improve the cytotoxic capacity of T cells [31,146]. However, blocking IFN-I during acute SIV infection in rhesus macaques resulted in the reduced expression of antiviral genes, increased size of the SIV reservoir, and accelerated CD4 T cell depletion and progression to AIDS [69]. In contrast, treatment with the IFN-1 antagonist in both ART-suppressed and ART-untreated chronically SIV-infected animals resulted in a reduction in IFN-I-mediated inflammation but had no effect on plasma viremia level [147].
Another approach to modulating excessive IFN-I signaling involves targeting the JAK-STAT pathway, which mediates the downstream effects of IFNAR activation. Small-molecule inhibitors of JAKs, such as Ruxolitinib, have shown effectiveness in reducing chronic inflammation and immune activation. In HIV research, in vitro studies have demonstrated that JAK inhibitors can reduce the frequency of cells harboring integrated HIV viral DNA and block the interleukin-15 (IL-15)-induced reactivation of latent HIV [148]. Additionally, a randomized trial indicated that Ruxolitinib significantly reduces immune activation in individuals living with HIV [149]. In the context of HIV, chronic IFN-I signaling not only impairs systemic immune responses but also plays a critical role in maintaining viral reservoirs and contributing to central nervous system (CNS) complications. Recent studies have also explored the potential of JAK inhibitors in mitigating the effects of HIV associated neurocognitive disorders (HANDs). In a severe combined immunodeficiency (SCID) mouse model intracranially injected with HIV-infected human monocyte-derived macrophages (MDMs), baricitinib, a JAK1/2 inhibitor, was shown to cross the blood–brain barrier and reverse cognitive deficits associated with HANDs [150]. By dampening chronic IFN-I signaling, Ruxolitinib treatment alleviated neuroinflammation, improved cognitive deficits, and reduced histopathological damage [151]. Those studies demonstrated that inhibiting the JAK/STAT signaling pathway could be a potential therapeutic strategy to finetuning type I IFN signaling for reducing HIV replication and managing HIV-related neurological complications.
Beyond direct inhibition of the IFN-I signaling pathway, modulating cellular processes that influence upstream innate sensing and downstream immune exhaustion offers an additional layer of therapeutic potential. Emerging evidence indicates that autophagy helps degrade cytosolic mitochondrial DNA (mtDNA) and viral nucleic acid–containing complexes, thereby limiting the chronic activation of cytosolic sensors such as cGAS-STING, which are key drivers of sustained IFN-I production [152,153,154]. In contrast, impaired autophagy leads to increased IFN-I responses [155,156,157]. Our recent study showed that the induction of autophagy with rapamycin reduced ISG expression, alleviated T cell exhaustion, and enhanced CD8⁺ T cell function in the setting of chronic HIV-induced immune dysfunction [156]. Taken together, these findings support a model in which targeting autophagy can fine-tune IFN-I signaling by limiting excessive innate immune activation at its source while simultaneously supporting T cell survival and function. Given its dual benefits, modulating autophagy to fine-tune type I IFN signaling may offer an alternative strategy to improve outcomes in chronic viral infection and immune exhaustion.
Considering IFN-I’s important role in innate immune responses during acute infection, the carefully timed and dosed administration of IFN-I can provide substantial benefits in viral infectious disease. This dichotomous nature of IFN-I signaling underscores the need for strategies that enhance its beneficial effects while minimizing its harmful consequences, particularly in the context of chronic inflammation and immune dysfunction. Recent research has shown that the timing and dosing of IFN-I administration or blocking are critical for IFN-I-based therapies [53,141,158]. During acute viral infections, the short-term administration of type I IFNs can amplify innate and adaptive immune responses, promoting rapid viral clearance in hepatitis C [53,159]. In HIV research, however, this concept has unique challenges due to the virus’s ability to persist and exploit immune activation. Early recombinant IFNα2a treatment in acute SIV infection enhanced antiviral responses, but prolonged use led to IFNα/β desensitization, increasing the SIV reservoir and CD4+ T cell loss [69]. In contrast, the blockade of IFNAR in combination with ART in HIV infected humanized mice facilitated faster viral suppression and reduced viral reservoir [31,146]. These findings underscore the critical role of timing in IFN-induced innate immune responses during acute SIV infection, suggesting that the timing of such responses is a key factor influencing disease progression, a concept likely applicable to various other infections.
Combination therapies involving IFN-I modulation offer a promising strategy for enhancing immune responses in both cancer and viral infections. Pairing IFN-I therapies with immune checkpoint inhibitors, such as anti-PD-1 or anti-CTLA-4 antibodies, has shown potential in reversing T cell exhaustion and improving the efficacy of immunotherapies. In cancer research, blocking IFNAR in combination with immune checkpoint inhibitors has been explored as a way to reinvigorate anti-tumor immunity [160]. Additionally, sustained type I IFN signaling has been identified as a mechanism of resistance to ICB, suggesting that modulating IFNAR activity could potentially overcome this resistance and improve therapeutic outcomes [161].
CAR-T cell therapy represents a promising “kill” component within the “shock and kill” strategy aimed at eradicating latent HIV reservoirs [162,163]. However, during chronic HIV infection, persistent IFN-I signaling and sustained immune activation drive CAR-T cell exhaustion, posing a major barrier to therapeutic success. Our recent study demonstrated that rapamycin, an mTOR inhibitor, can counteract these challenges by enhancing mitochondrial function, reducing chronic inflammation, and preventing CAR-T cell exhaustion [164]. These effects collectively support a more favorable immune environment for CAR-T activity, potentially increasing the efficiency of reservoir clearance. By alleviating IFN-I-driven immune dysfunction, rapamycin may help “recondition” the immune system, enabling more robust CAR-T cell responses in “shock and kill” approaches [156,164].
Targeting pDCs has also emerged as a strategic target for dampening the dysregulated type I IFN response in autoimmune diseases like SLE. Targeting ILT7 with the monoclonal antibody VIB7734 significantly reduced circulating and skin-resident pDCs in patients with cutaneous lupus erythematosus, leading to decreased local IFN-I activity and clinical improvement [165,166], while transient pDC depletion in a lupus-prone mouse model similarly diminished immune cell activation, autoantibody production, and kidney inflammation, underscoring the central role of pDC-driven IFN-I in lupus pathogenesis [167]. Translating this approach to HIV, where pDCs sustain a high level of IFN-I during chronic infection, is compelling. This mechanistic rationale highlights pDCs targeting as a potential adjunctive strategy in chronic HIV infection—by curtailing the chief source of IFN-I, it may be possible to break the cycle of IFN-driven inflammation and immune dysfunction, thereby bolstering host immunity and improving clinical outcomes [6].
The integration of these strategies—precisely timed IFN-I administration, combination therapies with checkpoint inhibitors, and metabolic enhancers—offers a powerful framework for improving immune responses in both virology and cancer. In the context of HIV, leveraging the antiviral properties of type I IFNs while minimizing their chronic effects has the potential to enhance curative strategies [26,168]. Future research will need to refine these approaches further, with an emphasis on understanding how to balance the pro- and anti-inflammatory effects of IFN-I signaling in different stages of infection or disease progression [169]. Another primary challenge lies in balancing efficacy and safety. Patient-specific factors, such as immune status, genetic predisposition, and underlying comorbidities should also be considered. Finetuning the delivery of IFN-I therapies to achieve the right balance between efficacy and safety will require extensive clinical trials and personalized treatment approaches.

6. Conclusions

Type I IFN signaling is a critical determinant of immune responses in HIV infection. While its early antiviral effects are indispensable, chronic dysregulation poses significant challenges to T cell immunity. Advances in our understanding of IFN-I signaling dynamics and the development of targeted therapeutic strategies offer hope for enhancing immune function and achieving better outcomes for individuals living with HIV. The future of IFN-I modulation lies in overcoming these challenges through innovative research and clinical advancements. Future research should focus on developing personalized treatment protocols that effectively balance the therapeutic benefits and potential risks of IFN modulation. In addition, developing robust biomarkers for patient stratification and treatment monitoring as well as unraveling the complex interactions between IFN-I and latent reservoirs are critical steps forward. Moreover, identifying the key ISGs that regulate type I IFN signaling during chronic infection could provide novel tools to fine-tune IFN responses at different stages of HIV infection. By addressing these challenges, IFN-I-based therapies have the potential to transform treatment paradigms for viral infections, bringing us closer to achieving long-lasting immune restoration and a functional cure for HIV infection.

Author Contributions

W.M. and A.Z. conceptualized this work. W.M., N.K. and A.Z. wrote this review together. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the National Institute of Allergy and Infectious Diseases (R01AI172727 to A.Z.), the National Institute on Drug Abuse (R01DA52841 and DA059873 to A.Z.), the American Foundation for AIDS Research (grants 110304-71-RKRL and 10395-72-RPRL to A.Z.), and the California HIV/AIDS Research Program (grant H24BD7817 to W.M.). Dr. Mu acknowledges the UCLA/CDU CFAR and UCLA AIDS Institute for seed grant support (Grant AI152501).

Data Availability Statement

No specific data were generated to support reported results.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. de Cock, K.M.; Jaffe, H.W.; Curran, J.W. The evolving epidemiology of HIV/AIDS. AIDS 2012, 26, 1205–1213. [Google Scholar] [CrossRef] [PubMed]
  2. Deeks, S.G.; Overbaugh, J.; Phillips, A.; Buchbinder, S. HIV infection. Nat. Rev. Dis. Primers 2015, 1, 15035. [Google Scholar] [CrossRef] [PubMed]
  3. Hatano, H. Immune activation and HIV persistence: Considerations for novel therapeutic interventions. Curr. Opin. HIV AIDS 2013, 8, 211–216. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  4. Cai, C.W.; Sereti, I. Residual immune dysfunction under antiretroviral therapy. Semin. Immunol. 2021, 51, 101471. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  5. Boyd, M.A.; van Bockel, D.; Munier, C.M.L.; Kelleher, A.D. Navigating the complexity of chronic HIV-1 associated immune dysregulation. Curr. Opin. Immunol. 2022, 76, 102186. [Google Scholar] [CrossRef] [PubMed]
  6. Su, L. Pathogenic Role of Type I Interferons in HIV-Induced Immune Impairments in Humanized Mice. Curr. HIV/AIDS Rep. 2019, 16, 224–229. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  7. Londe, A.C.; Fernandez-Ruiz, R.; Julio, P.R.; Appenzeller, S.; Niewold, T.B. Type I Interferons in Autoimmunity: Implications in Clinical Phenotypes and Treatment Response. J. Rheumatol. 2023, 50, 1103–1113. [Google Scholar] [CrossRef] [PubMed]
  8. Chen, K.; Liu, J.; Cao, X. Regulation of type I interferon signaling in immunity and inflammation: A comprehensive review. J. Autoimmun. 2017, 83, 1–11. [Google Scholar] [CrossRef] [PubMed]
  9. Schoggins, J.W.; Wilson, S.J.; Panis, M.; Murphy, M.Y.; Jones, C.T.; Bieniasz, P.; Rice, C.M. A diverse range of gene products are effectors of the type I interferon antiviral response. Nature 2011, 472, 481–485. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  10. Schoggins, J.W. Interferon-Stimulated Genes: What Do They All Do? Annu. Rev. Virol. 2019, 6, 567–584. [Google Scholar] [CrossRef] [PubMed]
  11. Ivashkiv, L.B.; Donlin, L.T. Regulation of type I interferon responses. Nat. Rev. Immunol. 2014, 14, 36–49. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  12. Bourdon, M.; Manet, C.; Montagutelli, X. Host genetic susceptibility to viral infections: The role of type I interferon induction. Genes. Immun. 2020, 21, 365–379. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  13. Li, D.; Wu, M. Pattern recognition receptors in health and diseases. Signal Transduct. Target. Ther. 2021, 6, 291. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  14. Smith, S.; Weston, S.; Kellam, P.; Marsh, M. IFITM proteins-cellular inhibitors of viral entry. Curr. Opin. Virol. 2014, 4, 71–77. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  15. Perng, Y.C.; Lenschow, D.J. ISG15 in antiviral immunity and beyond. Nat. Rev. Microbiol. 2018, 16, 423–439. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  16. Kristiansen, H.; Scherer, C.A.; McVean, M.; Iadonato, S.P.; Vends, S.; Thavachelvam, K.; Steffensen, T.B.; Horan, K.A.; Kuri, T.; Weber, F.; et al. Extracellular 2′-5′ oligoadenylate synthetase stimulates RNase L-independent antiviral activity: A novel mechanism of virus-induced innate immunity. J. Virol. 2010, 84, 11898–11904. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  17. Staeheli, P.; Haller, O. Human MX2/MxB: A Potent Interferon-Induced Postentry Inhibitor of Herpesviruses and HIV-1. J. Virol. 2018, 92, e00709-18. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  18. Yang, E.; Li, M.M.H. All About the RNA: Interferon-Stimulated Genes That Interfere With Viral RNA Processes. Front. Immunol. 2020, 11, 605024. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  19. Schoggins, J.W. Interferon-stimulated genes: Roles in viral pathogenesis. Curr. Opin. Virol. 2014, 6, 40–46. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  20. Sadler, A.J.; Williams, B.R. Interferon-inducible antiviral effectors. Nat. Rev. Immunol. 2008, 8, 559–568. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  21. Metz, P.; Reuter, A.; Bender, S.; Bartenschlager, R. Interferon-stimulated genes and their role in controlling hepatitis C virus. J. Hepatol. 2013, 59, 1331–1341. [Google Scholar] [CrossRef] [PubMed]
  22. Crosse, K.M.; Monson, E.A.; Beard, M.R.; Helbig, K.J. Interferon-Stimulated Genes as Enhancers of Antiviral Innate Immune Signaling. J. Innate Immun. 2018, 10, 85–93. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  23. Wilson, E.B.; Yamada, D.H.; Elsaesser, H.; Herskovitz, J.; Deng, J.; Cheng, G.; Aronow, B.J.; Karp, C.L.; Brooks, D.G. Blockade of chronic type I interferon signaling to control persistent LCMV infection. Science 2013, 340, 202–207. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  24. van Pul, L.; van Dort, K.A.; Girigorie, A.F.; Maurer, I.; Harskamp, A.M.; Kootstra, N.A. Human Immunodeficiency Virus-Induced Interferon-Stimulated Gene Expression Is Associated with Monocyte Activation and Predicts Viral Load. Open Forum Infect. Dis. 2024, 11, ofae434. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  25. Sugawara, S.; Thomas, D.L.; Balagopal, A. HIV-1 Infection and Type 1 Interferon: Navigating Through Uncertain Waters. AIDS Res. Hum. Retroviruses 2019, 35, 25–32. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  26. Scagnolari, C.; Antonelli, G. Type I interferon and HIV: Subtle balance between antiviral activity, immunopathogenesis and the microbiome. Cytokine Growth Factor. Rev. 2018, 40, 19–31. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  27. Murira, A.; Lamarre, A. Type-I Interferon Responses: From Friend to Foe in the Battle against Chronic Viral Infection. Front. Immunol. 2016, 7, 609. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  28. Dagenais-Lussier, X.; Loucif, H.; Murira, A.; Laulhe, X.; Stager, S.; Lamarre, A.; van Grevenynghe, J. Sustained IFN-I Expression during Established Persistent Viral Infection: A “Bad Seed” for Protective Immunity. Viruses 2017, 10, 12. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  29. Sumida, T.S.; Dulberg, S.; Schupp, J.C.; Lincoln, M.R.; Stillwell, H.A.; Axisa, P.P.; Comi, M.; Unterman, A.; Kaminski, N.; Madi, A.; et al. Type I interferon transcriptional network regulates expression of coinhibitory receptors in human T cells. Nat. Immunol. 2022, 23, 632–642. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  30. Chen, W.; Teo, J.M.N.; Yau, S.W.; Wong, M.Y.; Lok, C.N.; Che, C.M.; Javed, A.; Huang, Y.; Ma, S.; Ling, G.S. Chronic type I interferon signaling promotes lipid-peroxidation-driven terminal CD8(+) T cell exhaustion and curtails anti-PD-1 efficacy. Cell Rep. 2022, 41, 111647. [Google Scholar] [CrossRef] [PubMed]
  31. Zhen, A.; Rezek, V.; Youn, C.; Lam, B.; Chang, N.; Rick, J.; Carrillo, M.; Martin, H.; Kasparian, S.; Syed, P.; et al. Targeting type I interferon-mediated activation restores immune function in chronic HIV infection. J. Clin. Investig. 2017, 127, 260–268. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  32. Wang, Y.; Swiecki, M.; Cella, M.; Alber, G.; Schreiber, R.D.; Gilfillan, S.; Colonna, M. Timing and magnitude of type I interferon responses by distinct sensors impact CD8 T cell exhaustion and chronic viral infection. Cell Host Microbe 2012, 11, 631–642. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  33. Lukhele, S.; Boukhaled, G.M.; Brooks, D.G. Type I interferon signaling, regulation and gene stimulation in chronic virus infection. Semin. Immunol. 2019, 43, 101277. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  34. Greene, T.T.; Zuniga, E.I. Type I Interferon Induction and Exhaustion during Viral Infection: Plasmacytoid Dendritic Cells and Emerging COVID-19 Findings. Viruses 2021, 13, 1829. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  35. Wieser, V.; Adolph, T.E.; Grander, C.; Grabherr, F.; Enrich, B.; Moser, P.; Moschen, A.R.; Kaser, S.; Tilg, H. Adipose type I interferon signalling protects against metabolic dysfunction. Gut 2018, 67, 157–165. [Google Scholar] [CrossRef] [PubMed]
  36. Lercher, A.; Bhattacharya, A.; Popa, A.M.; Caldera, M.; Schlapansky, M.F.; Baazim, H.; Agerer, B.; Gurtl, B.; Kosack, L.; Majek, P.; et al. Type I Interferon Signaling Disrupts the Hepatic Urea Cycle and Alters Systemic Metabolism to Suppress T Cell Function. Immunity 2019, 51, 1074–1087.e1079. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  37. Jain, S.; Bemark, M.; Spencer, J. Human gut-associated lymphoid tissue: A dynamic hub propagating modulators of inflammation. Clin. Transl. Med. 2023, 13, e1417. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  38. Wang, B.; Kang, W.; Zuo, J.; Kang, W.; Sun, Y. The Significance of Type-I Interferons in the Pathogenesis and Therapy of Human Immunodeficiency Virus 1 Infection. Front. Immunol. 2017, 8, 1431. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  39. Vermeire, J.; Roesch, F.; Sauter, D.; Rua, R.; Hotter, D.; Van Nuffel, A.; Vanderstraeten, H.; Naessens, E.; Iannucci, V.; Landi, A.; et al. HIV Triggers a cGAS-Dependent, Vpu- and Vpr-Regulated Type I Interferon Response in CD4(+) T Cells. Cell Rep. 2016, 17, 413–424. [Google Scholar] [CrossRef] [PubMed]
  40. Lepelley, A.; Louis, S.; Sourisseau, M.; Law, H.K.; Pothlichet, J.; Schilte, C.; Chaperot, L.; Plumas, J.; Randall, R.E.; Si-Tahar, M.; et al. Innate sensing of HIV-infected cells. PLoS Pathog. 2011, 7, e1001284. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  41. Mazewski, C.; Perez, R.E.; Fish, E.N.; Platanias, L.C. Type I Interferon (IFN)-Regulated Activation of Canonical and Non-Canonical Signaling Pathways. Front. Immunol. 2020, 11, 606456. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  42. Yan, N.; Chen, Z.J. Intrinsic antiviral immunity. Nat. Immunol. 2012, 13, 214–222. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  43. Schneider, W.M.; Chevillotte, M.D.; Rice, C.M. Interferon-stimulated genes: A complex web of host defenses. Annu. Rev. Immunol. 2014, 32, 513–545. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  44. Neil, S.J.; Zang, T.; Bieniasz, P.D. Tetherin inhibits retrovirus release and is antagonized by HIV-1 Vpu. Nature 2008, 451, 425–430. [Google Scholar] [CrossRef] [PubMed]
  45. Goujon, C.; Malim, M.H. Characterization of the alpha interferon-induced postentry block to HIV-1 infection in primary human macrophages and T cells. J. Virol. 2010, 84, 9254–9266. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  46. Pery, E.; Sheehy, A.; Nebane, N.M.; Brazier, A.J.; Misra, V.; Rajendran, K.S.; Buhrlage, S.J.; Mankowski, M.K.; Rasmussen, L.; White, E.L.; et al. Identification of a novel HIV-1 inhibitor targeting Vif-dependent degradation of human APOBEC3G protein. J. Biol. Chem. 2015, 290, 10504–10517. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  47. Harris, R.S.; Liddament, M.T. Retroviral restriction by APOBEC proteins. Nat. Rev. Immunol. 2004, 4, 868–877. [Google Scholar] [CrossRef] [PubMed]
  48. Goila-Gaur, R.; Strebel, K. HIV-1 Vif, APOBEC, and intrinsic immunity. Retrovirology 2008, 5, 51. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  49. Laguette, N.; Sobhian, B.; Casartelli, N.; Ringeard, M.; Chable-Bessia, C.; Segeral, E.; Yatim, A.; Emiliani, S.; Schwartz, O.; Benkirane, M. SAMHD1 is the dendritic- and myeloid-cell-specific HIV-1 restriction factor counteracted by Vpx. Nature 2011, 474, 654–657. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  50. Goldstone, D.C.; Ennis-Adeniran, V.; Hedden, J.J.; Groom, H.C.; Rice, G.I.; Christodoulou, E.; Walker, P.A.; Kelly, G.; Haire, L.F.; Yap, M.W.; et al. HIV-1 restriction factor SAMHD1 is a deoxynucleoside triphosphate triphosphohydrolase. Nature 2011, 480, 379–382. [Google Scholar] [CrossRef] [PubMed]
  51. Martinez, J.; Huang, X.; Yang, Y. Direct action of type I IFN on NK cells is required for their activation in response to vaccinia viral infection in vivo. J. Immunol. 2008, 180, 1592–1597. [Google Scholar] [CrossRef] [PubMed]
  52. Kwaa, A.K.R.; Talana, C.A.G.; Blankson, J.N. Interferon Alpha Enhances NK Cell Function and the Suppressive Capacity of HIV-Specific CD8(+) T Cells. J. Virol. 2019, 93. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  53. McNab, F.; Mayer-Barber, K.; Sher, A.; Wack, A.; O’Garra, A. Type I interferons in infectious disease. Nat. Rev. Immunol. 2015, 15, 87–103. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  54. Madera, S.; Rapp, M.; Firth, M.A.; Beilke, J.N.; Lanier, L.L.; Sun, J.C. Type I IFN promotes NK cell expansion during viral infection by protecting NK cells against fratricide. J. Exp. Med. 2016, 213, 225–233. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  55. Parlato, S.; Santini, S.M.; Lapenta, C.; Di Pucchio, T.; Logozzi, M.; Spada, M.; Giammarioli, A.M.; Malorni, W.; Fais, S.; Belardelli, F. Expression of CCR-7, MIP-3beta, and Th-1 chemokines in type I IFN-induced monocyte-derived dendritic cells: Importance for the rapid acquisition of potent migratory and functional activities. Blood 2001, 98, 3022–3029. [Google Scholar] [CrossRef] [PubMed]
  56. Lapenta, C.; Santini, S.M.; Spada, M.; Donati, S.; Urbani, F.; Accapezzato, D.; Franceschini, D.; Andreotti, M.; Barnaba, V.; Belardelli, F. IFN-alpha-conditioned dendritic cells are highly efficient in inducing cross-priming CD8(+) T cells against exogenous viral antigens. Eur. J. Immunol. 2006, 36, 2046–2060. [Google Scholar] [CrossRef] [PubMed]
  57. Beignon, A.S.; McKenna, K.; Skoberne, M.; Manches, O.; DaSilva, I.; Kavanagh, D.G.; Larsson, M.; Gorelick, R.J.; Lifson, J.D.; Bhardwaj, N. Endocytosis of HIV-1 activates plasmacytoid dendritic cells via Toll-like receptor-viral RNA interactions. J. Clin. Investig. 2005, 115, 3265–3275. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  58. Villadangos, J.A.; Young, L. Antigen-presentation properties of plasmacytoid dendritic cells. Immunity 2008, 29, 352–361. [Google Scholar] [CrossRef] [PubMed]
  59. Santini, S.M.; Lapenta, C.; Logozzi, M.; Parlato, S.; Spada, M.; di Pucchio, T.; Belardelli, F. Type I interferon as a powerful adjuvant for monocyte-derived dendritic cell development and activity in vitro and in Hu-PBL-SCID mice. J. Exp. Med. 2000, 191, 1777–1788. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  60. Joffre, O.P.; Segura, E.; Savina, A.; Amigorena, S. Cross-presentation by dendritic cells. Nat. Rev. Immunol. 2012, 12, 557–569. [Google Scholar] [CrossRef] [PubMed]
  61. Stacey, A.R.; Norris, P.J.; Qin, L.; Haygreen, E.A.; Taylor, E.; Heitman, J.; Lebedeva, M.; DeCamp, A.; Li, D.; Grove, D.; et al. Induction of a striking systemic cytokine cascade prior to peak viremia in acute human immunodeficiency virus type 1 infection, in contrast to more modest and delayed responses in acute hepatitis B and C virus infections. J. Virol. 2009, 83, 3719–3733. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  62. Hardy, G.A.; Sieg, S.; Rodriguez, B.; Anthony, D.; Asaad, R.; Jiang, W.; Mudd, J.; Schacker, T.; Funderburg, N.T.; Pilch-Cooper, H.A.; et al. Interferon-alpha is the primary plasma type-I IFN in HIV-1 infection and correlates with immune activation and disease markers. PLoS ONE 2013, 8, e56527. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  63. Lane, H.C.; Davey, V.; Kovacs, J.A.; Feinberg, J.; Metcalf, J.A.; Herpin, B.; Walker, R.; Deyton, L.; Davey, R.T., Jr.; Falloon, J.; et al. Interferon-alpha in patients with asymptomatic human immunodeficiency virus (HIV) infection. A randomized, placebo-controlled trial. Ann. Intern. Med. 1990, 112, 805–811. [Google Scholar] [CrossRef] [PubMed]
  64. Cheng, L.; Yu, H.; Li, G.; Li, F.; Ma, J.; Li, J.; Chi, L.; Zhang, L.; Su, L. Type I interferons suppress viral replication but contribute to T cell depletion and dysfunction during chronic HIV-1 infection. JCI Insight 2017, 2, e94366. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  65. Doitsh, G.; Galloway, N.L.; Geng, X.; Yang, Z.; Monroe, K.M.; Zepeda, O.; Hunt, P.W.; Hatano, H.; Sowinski, S.; Munoz-Arias, I.; et al. Cell death by pyroptosis drives CD4 T-cell depletion in HIV-1 infection. Nature 2014, 505, 509–514. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  66. Bosinger, S.E.; Li, Q.; Gordon, S.N.; Klatt, N.R.; Duan, L.; Xu, L.; Francella, N.; Sidahmed, A.; Smith, A.J.; Cramer, E.M.; et al. Global genomic analysis reveals rapid control of a robust innate response in SIV-infected sooty mangabeys. J. Clin. Investig. 2009, 119, 3556–3572. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  67. Jacquelin, B.; Mayau, V.; Targat, B.; Liovat, A.S.; Kunkel, D.; Petitjean, G.; Dillies, M.A.; Roques, P.; Butor, C.; Silvestri, G.; et al. Nonpathogenic SIV infection of African green monkeys induces a strong but rapidly controlled type I IFN response. J. Clin. Investig. 2009, 119, 3544–3555. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  68. Lederer, S.; Favre, D.; Walters, K.A.; Proll, S.; Kanwar, B.; Kasakow, Z.; Baskin, C.R.; Palermo, R.; McCune, J.M.; Katze, M.G. Transcriptional profiling in pathogenic and non-pathogenic SIV infections reveals significant distinctions in kinetics and tissue compartmentalization. PLoS Pathog. 2009, 5, e1000296. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  69. Sandler, N.G.; Bosinger, S.E.; Estes, J.D.; Zhu, R.T.; Tharp, G.K.; Boritz, E.; Levin, D.; Wijeyesinghe, S.; Makamdop, K.N.; del Prete, G.Q.; et al. Type I interferon responses in rhesus macaques prevent SIV infection and slow disease progression. Nature 2014, 511, 601–605. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  70. Mandl, J.N.; Barry, A.P.; Vanderford, T.H.; Kozyr, N.; Chavan, R.; Klucking, S.; Barrat, F.J.; Coffman, R.L.; Staprans, S.I.; Feinberg, M.B. Divergent TLR7 and TLR9 signaling and type I interferon production distinguish pathogenic and nonpathogenic AIDS virus infections. Nat. Med. 2008, 14, 1077–1087. [Google Scholar] [CrossRef] [PubMed]
  71. Jasinska, A.J.; Apetrei, C.; Pandrea, I. Walk on the wild side: SIV infection in African non-human primate hosts-from the field to the laboratory. Front. Immunol. 2022, 13, 1060985. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  72. Harris, L.D.; Tabb, B.; Sodora, D.L.; Paiardini, M.; Klatt, N.R.; Douek, D.C.; Silvestri, G.; Muller-Trutwin, M.; Vasile-Pandrea, I.; Apetrei, C.; et al. Downregulation of robust acute type I interferon responses distinguishes nonpathogenic simian immunodeficiency virus (SIV) infection of natural hosts from pathogenic SIV infection of rhesus macaques. J. Virol. 2010, 84, 7886–7891. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  73. Sedaghat, A.R.; German, J.; Teslovich, T.M.; Cofrancesco, J., Jr.; Jie, C.C.; Talbot, C.C., Jr.; Siliciano, R.F. Chronic CD4+ T-cell activation and depletion in human immunodeficiency virus type 1 infection: Type I interferon-mediated disruption of T-cell dynamics. J. Virol. 2008, 82, 1870–1883. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  74. Hardy, G.A.; Sieg, S.F.; Rodriguez, B.; Jiang, W.; Asaad, R.; Lederman, M.M.; Harding, C.V. Desensitization to type I interferon in HIV-1 infection correlates with markers of immune activation and disease progression. Blood 2009, 113, 5497–5505. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  75. Scagnolari, C.; Monteleone, K.; Selvaggi, C.; Pierangeli, A.; D’Ettorre, G.; Mezzaroma, I.; Turriziani, O.; Gentile, M.; Vullo, V.; Antonelli, G. ISG15 expression correlates with HIV-1 viral load and with factors regulating T cell response. Immunobiology 2016, 221, 282–290. [Google Scholar] [CrossRef] [PubMed]
  76. Wang, X.M.; Zhang, J.Y.; Xing, X.; Huang, H.H.; Xia, P.; Dai, X.P.; Hu, W.; Zhang, C.; Song, J.W.; Fan, X.; et al. Global transcriptomic characterization of T cells in individuals with chronic HIV-1 infection. Cell Discov. 2022, 8, 29. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  77. Mackelprang, R.D.; Filali-Mouhim, A.; Richardson, B.; Lefebvre, F.; Katabira, E.; Ronald, A.; Gray, G.; Cohen, K.W.; Klatt, N.R.; Pecor, T.; et al. Upregulation of IFN-stimulated genes persists beyond the transitory broad immunologic changes of acute HIV-1 infection. iScience 2023, 26, 106454. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  78. Kahan, S.M.; Wherry, E.J.; Zajac, A.J. T cell exhaustion during persistent viral infections. Virology 2015, 479–480, 180–193. [Google Scholar] [CrossRef] [PubMed Central]
  79. Deeks, S.G.; Tracy, R.; Douek, D.C. Systemic effects of inflammation on health during chronic HIV infection. Immunity 2013, 39, 633–645. [Google Scholar] [CrossRef] [PubMed Central]
  80. Griesbeck, M.; Scully, E.; Altfeld, M. Sex and gender differences in HIV-1 infection. Clin. Sci. 2016, 130, 1435–1451. [Google Scholar] [CrossRef] [PubMed]
  81. Meier, A.; Chang, J.J.; Chan, E.S.; Pollard, R.B.; Sidhu, H.K.; Kulkarni, S.; Wen, T.F.; Lindsay, R.J.; Orellana, L.; Mildvan, D.; et al. Sex differences in the Toll-like receptor-mediated response of plasmacytoid dendritic cells to HIV-1. Nat. Med. 2009, 15, 955–959. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  82. Guery, J.C. Sex Differences in Primary HIV Infection: Revisiting the Role of TLR7-Driven Type 1 IFN Production by Plasmacytoid Dendritic Cells in Women. Front. Immunol. 2021, 12, 729233. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  83. Chang, J.J.; Woods, M.; Lindsay, R.J.; Doyle, E.H.; Griesbeck, M.; Chan, E.S.; Robbins, G.K.; Bosch, R.J.; Altfeld, M. Higher expression of several interferon-stimulated genes in HIV-1-infected females after adjusting for the level of viral replication. J. Infect. Dis. 2013, 208, 830–838. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  84. Sterling, T.R.; Lyles, C.M.; Vlahov, D.; Astemborski, J.; Margolick, J.B.; Quinn, T.C. Sex differences in longitudinal human immunodeficiency virus type 1 RNA levels among seroconverters. J. Infect. Dis. 1999, 180, 666–672. [Google Scholar] [CrossRef] [PubMed]
  85. Farzadegan, H.; Hoover, D.R.; Astemborski, J.; Lyles, C.M.; Margolick, J.B.; Markham, R.B.; Quinn, T.C.; Vlahov, D. Sex differences in HIV-1 viral load and progression to AIDS. Lancet 1998, 352, 1510–1514. [Google Scholar] [CrossRef] [PubMed]
  86. Terawaki, S.; Chikuma, S.; Shibayama, S.; Hayashi, T.; Yoshida, T.; Okazaki, T.; Honjo, T. IFN-alpha directly promotes programmed cell death-1 transcription and limits the duration of T cell-mediated immunity. J. Immunol. 2011, 186, 2772–2779. [Google Scholar] [CrossRef] [PubMed]
  87. Jones, R.B.; Ndhlovu, L.C.; Barbour, J.D.; Sheth, P.M.; Jha, A.R.; Long, B.R.; Wong, J.C.; Satkunarajah, M.; Schweneker, M.; Chapman, J.M.; et al. Tim-3 expression defines a novel population of dysfunctional T cells with highly elevated frequencies in progressive HIV-1 infection. J. Exp. Med. 2008, 205, 2763–2779. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  88. Saeidi, A.; Zandi, K.; Cheok, Y.Y.; Saeidi, H.; Wong, W.F.; Lee, C.Y.Q.; Cheong, H.C.; Yong, Y.K.; Larsson, M.; Shankar, E.M. T-Cell Exhaustion in Chronic Infections: Reversing the State of Exhaustion and Reinvigorating Optimal Protective Immune Responses. Front. Immunol. 2018, 9, 2569. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  89. Gao, Z.; Feng, Y.; Xu, J.; Liang, J. T-cell exhaustion in immune-mediated inflammatory diseases: New implications for immunotherapy. Front. Immunol. 2022, 13, 977394. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  90. Mu, W.; Patankar, V.; Kitchen, S.; Zhen, A. Examining Chronic Inflammation, Immune Metabolism, and T Cell Dysfunction in HIV Infection. Viruses 2024, 16, 219. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  91. Fritsch, S.D.; Weichhart, T. Effects of Interferons and Viruses on Metabolism. Front. Immunol. 2016, 7, 630. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  92. Brooks, D.G.; Walsh, K.B.; Elsaesser, H.; Oldstone, M.B. IL-10 directly suppresses CD4 but not CD8 T cell effector and memory responses following acute viral infection. Proc. Natl. Acad. Sci. USA 2010, 107, 3018–3023. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  93. Brooks, D.G.; Trifilo, M.J.; Edelmann, K.H.; Teyton, L.; McGavern, D.B.; Oldstone, M.B. Interleukin-10 determines viral clearance or persistence in vivo. Nat. Med. 2006, 12, 1301–1309. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  94. Boettler, T.; Cheng, Y.; Ehrhardt, K.; von Herrath, M. TGF-beta blockade does not improve control of an established persistent viral infection. Viral Immunol. 2012, 25, 232–238. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  95. Fraietta, J.A.; Mueller, Y.M.; Yang, G.; Boesteanu, A.C.; Gracias, D.T.; Do, D.H.; Hope, J.L.; Kathuria, N.; McGettigan, S.E.; Lewis, M.G.; et al. Type I interferon upregulates Bak and contributes to T cell loss during human immunodeficiency virus (HIV) infection. PLoS Pathog. 2013, 9, e1003658. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  96. Fernandez, S.; Tanaskovic, S.; Helbig, K.; Rajasuriar, R.; Kramski, M.; Murray, J.M.; Beard, M.; Purcell, D.; Lewin, S.R.; Price, P.; et al. CD4+ T-cell deficiency in HIV patients responding to antiretroviral therapy is associated with increased expression of interferon-stimulated genes in CD4+ T cells. J. Infect. Dis. 2011, 204, 1927–1935. [Google Scholar] [CrossRef] [PubMed]
  97. Kwa, S.; Kannanganat, S.; Nigam, P.; Siddiqui, M.; Shetty, R.D.; Armstrong, W.; Ansari, A.; Bosinger, S.E.; Silvestri, G.; Amara, R.R. Plasmacytoid dendritic cells are recruited to the colorectum and contribute to immune activation during pathogenic SIV infection in rhesus macaques. Blood 2011, 118, 2763–2773. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  98. Zeng, M.; Southern, P.J.; Reilly, C.S.; Beilman, G.J.; Chipman, J.G.; Schacker, T.W.; Haase, A.T. Lymphoid tissue damage in HIV-1 infection depletes naive T cells and limits T cell reconstitution after antiretroviral therapy. PLoS Pathog. 2012, 8, e1002437. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  99. Schacker, T.W.; Nguyen, P.L.; Beilman, G.J.; Wolinsky, S.; Larson, M.; Reilly, C.; Haase, A.T. Collagen deposition in HIV-1 infected lymphatic tissues and T cell homeostasis. J. Clin. Investig. 2002, 110, 1133–1139. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  100. Estes, J.D. Pathobiology of HIV/SIV-associated changes in secondary lymphoid tissues. Immunol. Rev. 2013, 254, 65–77. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  101. Ahodantin, J.; Nio, K.; Funaki, M.; Zhai, X.; Wilson, E.; Kottilil, S.; Cheng, L.; Li, G.; Su, L. Type I interferons and TGF-beta cooperate to induce liver fibrosis during HIV-1 infection under antiretroviral therapy. JCI Insight 2022, 7, e152738. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  102. Boulware, D.R.; Hullsiek, K.H.; Puronen, C.E.; Rupert, A.; Baker, J.V.; French, M.A.; Bohjanen, P.R.; Novak, R.M.; Neaton, J.D.; Sereti, I.; et al. Higher levels of CRP, D-dimer, IL-6, and hyaluronic acid before initiation of antiretroviral therapy (ART) are associated with increased risk of AIDS or death. J. Infect. Dis. 2011, 203, 1637–1646. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  103. Brenchley, J.M.; Price, D.A.; Schacker, T.W.; Asher, T.E.; Silvestri, G.; Rao, S.; Kazzaz, Z.; Bornstein, E.; Lambotte, O.; Altmann, D.; et al. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat. Med. 2006, 12, 1365–1371. [Google Scholar] [CrossRef] [PubMed]
  104. Hunt, P.W.; Lee, S.A.; Siedner, M.J. Immunologic Biomarkers, Morbidity, and Mortality in Treated HIV Infection. J. Infect. Dis. 2016, 214 (Suppl. S2), S44–S50. [Google Scholar] [CrossRef] [PubMed Central]
  105. Swainson, L.A.; Sharma, A.A.; Ghneim, K.; Ribeiro, S.P.; Wilkinson, P.; Dunham, R.M.; Albright, R.G.; Wong, S.; Estes, J.D.; Piatak, M.; et al. IFN-alpha blockade during ART-treated SIV infection lowers tissue vDNA, rescues immune function, and improves overall health. JCI Insight 2022, 7, e153046. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  106. Pantel, A.; Teixeira, A.; Haddad, E.; Wood, E.G.; Steinman, R.M.; Longhi, M.P. Direct type I IFN but not MDA5/TLR3 activation of dendritic cells is required for maturation and metabolic shift to glycolysis after poly IC stimulation. PLoS Biol. 2014, 12, e1001759. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  107. Hedl, M.; Yan, J.; Abraham, C. IRF5 and IRF5 Disease-Risk Variants Increase Glycolysis and Human M1 Macrophage Polarization by Regulating Proximal Signaling and Akt2 Activation. Cell Rep. 2016, 16, 2442–2455. [Google Scholar] [CrossRef] [PubMed Central]
  108. Burke, J.D.; Platanias, L.C.; Fish, E.N. Beta interferon regulation of glucose metabolism is PI3K/Akt dependent and important for antiviral activity against coxsackievirus B3. J. Virol. 2014, 88, 3485–3495. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  109. Wu, M.; Neilson, A.; Swift, A.L.; Moran, R.; Tamagnine, J.; Parslow, D.; Armistead, S.; Lemire, K.; Orrell, J.; Teich, J.; et al. Multiparameter metabolic analysis reveals a close link between attenuated mitochondrial bioenergetic function and enhanced glycolysis dependency in human tumor cells. Am. J. Physiol. Cell Physiol. 2007, 292, C125–C136. [Google Scholar] [CrossRef] [PubMed]
  110. Cao, J.; Liao, S.; Zeng, F.; Liao, Q.; Luo, G.; Zhou, Y. Effects of altered glycolysis levels on CD8(+) T cell activation and function. Cell Death Dis. 2023, 14, 407. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  111. Lewis, J.A.; Huq, A.; Najarro, P. Inhibition of mitochondrial function by interferon. J. Biol. Chem. 1996, 271, 13184–13190. [Google Scholar] [CrossRef] [PubMed]
  112. Haghikia, A.; Faissner, S.; Pappas, D.; Pula, B.; Akkad, D.A.; Arning, L.; Ruhrmann, S.; Duscha, A.; Gold, R.; Baranzini, S.E.; et al. Interferon-beta affects mitochondrial activity in CD4+ lymphocytes: Implications for mechanism of action in multiple sclerosis. Mult. Scler. 2015, 21, 1262–1270. [Google Scholar] [CrossRef] [PubMed]
  113. Olson, G.S.; Murray, T.A.; Jahn, A.N.; Mai, D.; Diercks, A.H.; Gold, E.S.; Aderem, A. Type I interferon decreases macrophage energy metabolism during mycobacterial infection. Cell Rep. 2021, 35, 109195. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  114. Radziszewska, A.; Peckham, H.; Restuadi, R.; Kartawinata, M.; Moulding, D.; de Gruijter, N.M.; Robinson, G.A.; Butt, M.; Deakin, C.T.; Wilkinson, M.G.L.; et al. Type I interferon and mitochondrial dysfunction are associated with dysregulated cytotoxic CD8+ T cell responses in juvenile systemic lupus erythematosus. Clin. Exp. Immunol. 2025, 219, uxae127. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  115. Yim, H.Y.; Yang, Y.; Lim, J.S.; Lee, M.S.; Zhang, D.E.; Kim, K.I. The mitochondrial pathway and reactive oxygen species are critical contributors to interferon-alpha/beta-mediated apoptosis in Ubp43-deficient hematopoietic cells. Biochem. Biophys. Res. Commun. 2012, 423, 436–440. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  116. Boasso, A. Type I Interferon at the Interface of Antiviral Immunity and Immune Regulation: The Curious Case of HIV-1. Scientifica 2013, 2013, 580968. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  117. Nguyen, K.B.; Watford, W.T.; Salomon, R.; Hofmann, S.R.; Pien, G.C.; Morinobu, A.; Gadina, M.; O’Shea, J.J.; Biron, C.A. Critical role for STAT4 activation by type 1 interferons in the interferon-gamma response to viral infection. Science 2002, 297, 2063–2066. [Google Scholar] [CrossRef] [PubMed]
  118. Belardelli, F. Role of interferons and other cytokines in the regulation of the immune response. APMIS 1995, 103, 161–179. [Google Scholar] [CrossRef] [PubMed]
  119. Castaldello, A.; Sgarbanti, M.; Marsili, G.; Brocca-Cofano, E.; Remoli, A.L.; Caputo, A.; Battistini, A. Interferon regulatory factor-1 acts as a powerful adjuvant in tat DNA based vaccination. J. Cell Physiol. 2010, 224, 702–709. [Google Scholar] [CrossRef] [PubMed]
  120. Longhi, M.P.; Trumpfheller, C.; Idoyaga, J.; Caskey, M.; Matos, I.; Kluger, C.; Salazar, A.M.; Colonna, M.; Steinman, R.M. Dendritic cells require a systemic type I interferon response to mature and induce CD4+ Th1 immunity with poly IC as adjuvant. J. Exp. Med. 2009, 206, 1589–1602. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  121. Curtsinger, J.M.; Valenzuela, J.O.; Agarwal, P.; Lins, D.; Mescher, M.F. Type I IFNs provide a third signal to CD8 T cells to stimulate clonal expansion and differentiation. J. Immunol. 2005, 174, 4465–4469. [Google Scholar] [CrossRef] [PubMed]
  122. Ito, T.; Amakawa, R.; Inaba, M.; Ikehara, S.; Inaba, K.; Fukuhara, S. Differential regulation of human blood dendritic cell subsets by IFNs. J. Immunol. 2001, 166, 2961–2969. [Google Scholar] [CrossRef] [PubMed]
  123. Akdis, C.A.; Blaser, K. Mechanisms of interleukin-10-mediated immune suppression. Immunology 2001, 103, 131–136. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  124. Mittal, S.K.; Roche, P.A. Suppression of antigen presentation by IL-10. Curr. Opin. Immunol. 2015, 34, 22–27. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  125. Yoshimura, A.; Wakabayashi, Y.; Mori, T. Cellular and molecular basis for the regulation of inflammation by TGF-beta. J. Biochem. 2010, 147, 781–792. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  126. Wan, Y.Y.; Flavell, R.A. ‘Yin-Yang’ functions of transforming growth factor-beta and T regulatory cells in immune regulation. Immunol. Rev. 2007, 220, 199–213. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  127. Li, M.O.; Wan, Y.Y.; Sanjabi, S.; Robertson, A.K.; Flavell, R.A. Transforming growth factor-beta regulation of immune responses. Annu. Rev. Immunol. 2006, 24, 99–146. [Google Scholar] [CrossRef] [PubMed]
  128. Josefowicz, S.Z.; Lu, L.F.; Rudensky, A.Y. Regulatory T cells: Mechanisms of differentiation and function. Annu. Rev. Immunol. 2012, 30, 531–564. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  129. Vandergeeten, C.; Fromentin, R.; Chomont, N. The role of cytokines in the establishment, persistence and eradication of the HIV reservoir. Cytokine Growth Factor. Rev. 2012, 23, 143–149. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  130. Beltra, J.C.; Decaluwe, H. Cytokines and persistent viral infections. Cytokine 2016, 82, 4–15. [Google Scholar] [CrossRef] [PubMed]
  131. Taleb, K.; Auffray, C.; Villefroy, P.; Pereira, A.; Hosmalin, A.; Gaudry, M.; Le Bon, A. Chronic Type I IFN Is Sufficient To Promote Immunosuppression through Accumulation of Myeloid-Derived Suppressor Cells. J. Immunol. 2017, 198, 1156–1163. [Google Scholar] [CrossRef] [PubMed]
  132. Cunningham, C.R.; Champhekar, A.; Tullius, M.V.; Dillon, B.J.; Zhen, A.; de la Fuente, J.R.; Herskovitz, J.; Elsaesser, H.; Snell, L.M.; Wilson, E.B.; et al. Type I and Type II Interferon Coordinately Regulate Suppressive Dendritic Cell Fate and Function during Viral Persistence. PLoS Pathog. 2016, 12, e1005356. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  133. Peyraud, F.; Guegan, J.P.; Bodet, D.; Cousin, S.; Bessede, A.; Italiano, A. Targeting Tryptophan Catabolism in Cancer Immunotherapy Era: Challenges and Perspectives. Front. Immunol. 2022, 13, 807271. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  134. Thompson, C.G.; Gay, C.L.; Kashuba, A.D.M. HIV Persistence in Gut-Associated Lymphoid Tissues: Pharmacological Challenges and Opportunities. AIDS Res. Hum. Retroviruses 2017, 33, 513–523. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  135. Samal, J.; Kelly, S.; Na-Shatal, A.; Elhakiem, A.; Das, A.; Ding, M.; Sanyal, A.; Gupta, P.; Melody, K.; Roland, B.; et al. Human immunodeficiency virus infection induces lymphoid fibrosis in the BM-liver-thymus-spleen humanized mouse model. JCI Insight 2018, 3, e120430. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  136. Estes, J.D.; Wietgrefe, S.; Schacker, T.; Southern, P.; Beilman, G.; Reilly, C.; Milush, J.M.; Lifson, J.D.; Sodora, D.L.; Carlis, J.V.; et al. Simian immunodeficiency virus-induced lymphatic tissue fibrosis is mediated by transforming growth factor beta 1-positive regulatory T cells and begins in early infection. J. Infect. Dis. 2007, 195, 551–561. [Google Scholar] [CrossRef] [PubMed]
  137. Huot, N.; Bosinger, S.E.; Paiardini, M.; Reeves, R.K.; Muller-Trutwin, M. Lymph Node Cellular and Viral Dynamics in Natural Hosts and Impact for HIV Cure Strategies. Front. Immunol. 2018, 9, 780. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  138. Zeng, M.; Smith, A.J.; Wietgrefe, S.W.; Southern, P.J.; Schacker, T.W.; Reilly, C.S.; Estes, J.D.; Burton, G.F.; Silvestri, G.; Lifson, J.D.; et al. Cumulative mechanisms of lymphoid tissue fibrosis and T cell depletion in HIV-1 and SIV infections. J. Clin. Investig. 2011, 121, 998–1008. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  139. Park, S.H.; Kang, K.; Giannopoulou, E.; Qiao, Y.; Kang, K.; Kim, G.; Park-Min, K.H.; Ivashkiv, L.B. Type I interferons and the cytokine TNF cooperatively reprogram the macrophage epigenome to promote inflammatory activation. Nat. Immunol. 2017, 18, 1104–1116. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  140. Kamada, R.; Yang, W.; Zhang, Y.; Patel, M.C.; Yang, Y.; Ouda, R.; Dey, A.; Wakabayashi, Y.; Sakaguchi, K.; Fujita, T.; et al. Interferon stimulation creates chromatin marks and establishes transcriptional memory. Proc. Natl. Acad. Sci. USA 2018, 115, E9162–E9171. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  141. Barrat, F.J.; Crow, M.K.; Ivashkiv, L.B. Interferon target-gene expression and epigenomic signatures in health and disease. Nat. Immunol. 2019, 20, 1574–1583. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  142. Mehta, S.; Jeffrey, K.L. Beyond receptors and signaling: Epigenetic factors in the regulation of innate immunity. Immunol. Cell Biol. 2015, 93, 233–244. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  143. Tausendschon, M.; Dehne, N.; Brune, B. Hypoxia causes epigenetic gene regulation in macrophages by attenuating Jumonji histone demethylase activity. Cytokine 2011, 53, 256–262. [Google Scholar] [CrossRef] [PubMed]
  144. Teijaro, J.R.; Ng, C.; Lee, A.M.; Sullivan, B.M.; Sheehan, K.C.; Welch, M.; Schreiber, R.D.; de la Torre, J.C.; Oldstone, M.B. Persistent LCMV infection is controlled by blockade of type I interferon signaling. Science 2013, 340, 207–211. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  145. Ng, C.T.; Sullivan, B.M.; Teijaro, J.R.; Lee, A.M.; Welch, M.; Rice, S.; Sheehan, K.C.; Schreiber, R.D.; Oldstone, M.B. Blockade of interferon Beta, but not interferon alpha, signaling controls persistent viral infection. Cell Host Microbe 2015, 17, 653–661. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  146. Cheng, L.; Ma, J.; Li, J.; Li, D.; Li, G.; Li, F.; Zhang, Q.; Yu, H.; Yasui, F.; Ye, C.; et al. Blocking type I interferon signaling enhances T cell recovery and reduces HIV-1 reservoirs. J. Clin. Investig. 2017, 127, 269–279. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  147. Nganou-Makamdop, K.; Billingsley, J.M.; Yaffe, Z.; O’Connor, G.; Tharp, G.K.; Ransier, A.; Laboune, F.; Matus-Nicodemos, R.; Lerner, A.; Gharu, L.; et al. Type I IFN signaling blockade by a PASylated antagonist during chronic SIV infection suppresses specific inflammatory pathways but does not alter T cell activation or virus replication. PLoS Pathog. 2018, 14, e1007246. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  148. Gavegnano, C.; Brehm, J.H.; Dupuy, F.P.; Talla, A.; Ribeiro, S.P.; Kulpa, D.A.; Cameron, C.; Santos, S.; Hurwitz, S.J.; Marconi, V.C.; et al. Novel mechanisms to inhibit HIV reservoir seeding using Jak inhibitors. PLoS Pathog. 2017, 13, e1006740. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  149. Marconi, V.C.; Moser, C.; Gavegnano, C.; Deeks, S.G.; Lederman, M.M.; Overton, E.T.; Tsibris, A.; Hunt, P.W.; Kantor, A.; Sekaly, R.P.; et al. Randomized Trial of Ruxolitinib in Antiretroviral-Treated Adults With Human Immunodeficiency Virus. Clin. Infect. Dis. 2022, 74, 95–104. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  150. Gavegnano, C.; Haile, W.B.; Hurwitz, S.; Tao, S.; Jiang, Y.; Schinazi, R.F.; Tyor, W.R. Baricitinib reverses HIV-associated neurocognitive disorders in a SCID mouse model and reservoir seeding in vitro. J. Neuroinflamm. 2019, 16, 182. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  151. Haile, W.B.; Gavegnano, C.; Tao, S.; Jiang, Y.; Schinazi, R.F.; Tyor, W.R. The Janus kinase inhibitor ruxolitinib reduces HIV replication in human macrophages and ameliorates HIV encephalitis in a murine model. Neurobiol. Dis. 2016, 92, 137–143. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  152. Prabakaran, T.; Bodda, C.; Krapp, C.; Zhang, B.C.; Christensen, M.H.; Sun, C.; Reinert, L.; Cai, Y.; Jensen, S.B.; Skouboe, M.K.; et al. Attenuation of cGAS-STING signaling is mediated by a p62/SQSTM1-dependent autophagy pathway activated by TBK1. EMBO J. 2018, 37, e97858. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  153. Liang, Q.; Seo, G.J.; Choi, Y.J.; Kwak, M.J.; Ge, J.; Rodgers, M.A.; Shi, M.; Leslie, B.J.; Hopfner, K.P.; Ha, T.; et al. Crosstalk between the cGAS DNA sensor and Beclin-1 autophagy protein shapes innate antimicrobial immune responses. Cell Host Microbe 2014, 15, 228–238. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  154. Gui, X.; Yang, H.; Li, T.; Tan, X.; Shi, P.; Li, M.; Du, F.; Chen, Z.J. Autophagy induction via STING trafficking is a primordial function of the cGAS pathway. Nature 2019, 567, 262–266. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  155. Perot, B.P.; Boussier, J.; Yatim, N.; Rossman, J.S.; Ingersoll, M.A.; Albert, M.L. Autophagy diminishes the early interferon-beta response to influenza A virus resulting in differential expression of interferon-stimulated genes. Cell Death Dis. 2018, 9, 539. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  156. Mu, W.; Rezek, V.; Martin, H.; Carrillo, M.A.; Tomer, S.; Hamid, P.; Lizarraga, M.A.; Tibbe, T.D.; Yang, O.O.; Jamieson, B.D.; et al. Autophagy inducer rapamycin treatment reduces IFN-I-mediated Inflammation and improves anti-HIV-1 T cell response in vivo. JCI Insight 2022, 7, e159136. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  157. Jounai, N.; Takeshita, F.; Kobiyama, K.; Sawano, A.; Miyawaki, A.; Xin, K.Q.; Ishii, K.J.; Kawai, T.; Akira, S.; Suzuki, K.; et al. The Atg5 Atg12 conjugate associates with innate antiviral immune responses. Proc. Natl. Acad. Sci. USA 2007, 104, 14050–14055. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  158. Snell, L.M.; McGaha, T.L.; Brooks, D.G. Type I Interferon in Chronic Virus Infection and Cancer. Trends Immunol. 2017, 38, 542–557. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  159. Badr, G.; Bedard, N.; Abdel-Hakeem, M.S.; Trautmann, L.; Willems, B.; Villeneuve, J.P.; Haddad, E.K.; Sekaly, R.P.; Bruneau, J.; Shoukry, N.H. Early interferon therapy for hepatitis C virus infection rescues polyfunctional, long-lived CD8+ memory T cells. J. Virol. 2008, 82, 10017–10031. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  160. Holzgruber, J.; Martins, C.; Kulcsar, Z.; Duplaine, A.; Rasbach, E.; Migayron, L.; Singh, P.; Statham, E.; Landsberg, J.; Boniface, K.; et al. Type I interferon signaling induces melanoma cell-intrinsic PD-1 and its inhibition antagonizes immune checkpoint blockade. Nat. Commun. 2024, 15, 7165. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  161. Jacquelot, N.; Yamazaki, T.; Roberti, M.P.; Duong, C.P.M.; Andrews, M.C.; Verlingue, L.; Ferrere, G.; Becharef, S.; Vetizou, M.; Daillere, R.; et al. Sustained Type I interferon signaling as a mechanism of resistance to PD-1 blockade. Cell Res. 2019, 29, 846–861. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  162. York, J.; Gowrishankar, K.; Micklethwaite, K.; Palmer, S.; Cunningham, A.L.; Nasr, N. Evolving Strategies to Eliminate the CD4 T Cells HIV Viral Reservoir via CAR T Cell Immunotherapy. Front. Immunol. 2022, 13, 873701. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  163. Mu, W.; Carrillo, M.A.; Kitchen, S.G. Engineering CAR T Cells to Target the HIV Reservoir. Front. Cell Infect. Microbiol. 2020, 10, 410. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  164. Mu, W.; Tomer, S.; Harding, J.; Kedia, N.; Rezek, V.; Cook, E.; Patankar, V.; Carrillo, M.A.; Martin, H.; Ng, H.; et al. Rapamycin enhances CAR-T control of HIV replication and reservoir elimination in vivo. J. Clin. Investig. 2025, 135, e185489. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  165. Ngo, C.; Garrec, C.; Tomasello, E.; Dalod, M. The role of plasmacytoid dendritic cells (pDCs) in immunity during viral infections and beyond. Cell Mol. Immunol. 2024, 21, 1008–1035. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  166. Karnell, J.L.; Wu, Y.; Mittereder, N.; Smith, M.A.; Gunsior, M.; Yan, L.; Casey, K.A.; Henault, J.; Riggs, J.M.; Nicholson, S.M.; et al. Depleting plasmacytoid dendritic cells reduces local type I interferon responses and disease activity in patients with cutaneous lupus. Sci. Transl. Med. 2021, 13, eabf8442. [Google Scholar] [CrossRef] [PubMed]
  167. Rowland, S.L.; Riggs, J.M.; Gilfillan, S.; Bugatti, M.; Vermi, W.; Kolbeck, R.; Unanue, E.R.; Sanjuan, M.A.; Colonna, M. Early, transient depletion of plasmacytoid dendritic cells ameliorates autoimmunity in a lupus model. J. Exp. Med. 2014, 211, 1977–1991. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  168. Harper, M.S.; Guo, K.; Gibbert, K.; Lee, E.J.; Dillon, S.M.; Barrett, B.S.; McCarter, M.D.; Hasenkrug, K.J.; Dittmer, U.; Wilson, C.C.; et al. Interferon-alpha Subtypes in an Ex Vivo Model of Acute HIV-1 Infection: Expression, Potency and Effector Mechanisms. PLoS Pathog. 2015, 11, e1005254. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  169. Osuji, F.N.; Onyenekwe, C.C.; Ahaneku, J.E.; Ukibe, N.R. The effects of highly active antiretroviral therapy on the serum levels of pro-inflammatory and anti-inflammatory cytokines in HIV infected subjects. J. Biomed. Sci. 2018, 25, 88. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Mu, W.; Kedia, N.; Zhen, A. Finetuning Type I Interferon Signaling to Enhance T Cell Immunity in HIV Infection. Viruses 2025, 17, 774. https://doi.org/10.3390/v17060774

AMA Style

Mu W, Kedia N, Zhen A. Finetuning Type I Interferon Signaling to Enhance T Cell Immunity in HIV Infection. Viruses. 2025; 17(6):774. https://doi.org/10.3390/v17060774

Chicago/Turabian Style

Mu, Wenli, Nandita Kedia, and Anjie Zhen. 2025. "Finetuning Type I Interferon Signaling to Enhance T Cell Immunity in HIV Infection" Viruses 17, no. 6: 774. https://doi.org/10.3390/v17060774

APA Style

Mu, W., Kedia, N., & Zhen, A. (2025). Finetuning Type I Interferon Signaling to Enhance T Cell Immunity in HIV Infection. Viruses, 17(6), 774. https://doi.org/10.3390/v17060774

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop