Fact-checked by Grok 2 weeks ago

Cross-presentation

Cross-presentation is a specialized process in which professional antigen-presenting cells, particularly dendritic cells (DCs), internalize exogenous antigens—such as those derived from pathogens, tumors, or apoptotic cells—and load them onto class I (MHC I) molecules for to + T cells, thereby initiating cytotoxic T responses. This mechanism contrasts with classical MHC I , which typically involves endogenous antigens synthesized within the presenting cell, and enables immune surveillance of threats that do not directly infect antigen-presenting cells. The importance of cross-presentation lies in its role in bridging innate and adaptive immunity, facilitating + T cell priming against intracellular pathogens like viruses, as well as tumors and transplants, while also contributing to and preventing under steady-state conditions. Primarily mediated by specialized subsets—such as XCR1+ conventional DCs (cDC1s, including CD8α+ or CD103+ in mice and CD141+ in humans)—cross-presentation is more efficient for particulate antigens (e.g., from ) than soluble ones, and its dysregulation can impair antitumor immunity or efficacy. Mechanistically, cross-presentation occurs via two main pathways: the vacuolar (or endosomal) pathway, where antigen processing and MHC I loading happen within phagosomes or endosomes independently of proteasomes and TAP transporters, involving lysosomal proteases like cathepsin S; and the cytosolic pathway, where antigens are exported from endosomes to the cytosol for proteasomal degradation, followed by peptide transport via TAP into the endoplasmic reticulum (ER) or endosomes for loading onto recycled MHC I molecules. Key regulators include Toll-like receptor (TLR) signaling, which modulates phagosomal maturation and ER-phagosome fusion via proteins like Sec22b, and components of the ER-associated degradation (ERAD) machinery, such as Sec61 and p97, that facilitate antigen translocation. Additional processes, like delayed antigen degradation in DCs and alternative routes such as gap junctions or cross-dressing, further enhance efficiency in specific contexts.

Introduction

Definition and Process Overview

Cross-presentation is the process by which professional antigen-presenting cells (APCs), particularly dendritic cells, internalize exogenous antigens from the extracellular environment, process them into peptides, and load these peptides onto class I (MHC I) molecules for presentation on the cell surface, thereby enabling recognition and activation by CD8+ T cells. This allows APCs to display antigens that originate outside the cell, such as those derived from pathogens, apoptotic cells, or tumors, which would otherwise not be accessible to the conventional MHC I pathway restricted to endogenous antigens. The general process begins with antigen uptake through mechanisms including phagocytosis, endocytosis, or receptor-mediated endocytosis, followed by transport to specialized intracellular compartments for processing. In these compartments, the antigens are degraded into peptides, which are then transported and loaded onto recycled or newly synthesized MHC I molecules. The peptide-MHC I complexes are subsequently transported to the cell surface, where they are recognized by cytotoxic T lymphocytes (CTLs), priming these cells for effector functions such as target cell lysis. Biologically, cross-presentation plays a pivotal role in immune surveillance by allowing APCs to detect and respond to infections or malignancies in cells that do not directly present antigens to the , such as non-hematopoietic cells harboring viruses or tumors. A key feature is the diversion of exogenous antigens, which are typically processed through the pathway for + T cell activation, into the MHC I pathway to elicit cytotoxic responses. This process is essential for initiating anti-viral and anti-tumor immunity, as well as maintaining to self-antigens.

Distinction from Conventional Antigen Presentation

Conventional MHC class I antigen presentation processes endogenous antigens, such as cytosolic proteins derived from intracellular pathogens like viruses or altered self-proteins, primarily within the cell's . These antigens are ubiquitinated and degraded into peptides by the , a multicatalytic complex. The resulting peptides, typically 8-10 in length, are then transported from the into the () lumen via the transporter associated with antigen processing (), a member of the ATP-binding cassette family. In the , these peptides are loaded onto newly synthesized MHC class I molecules, which consist of a heavy chain, β2-microglobulin, and chaperones like and tapasin for stable assembly; the peptide-MHC I complexes then traffic through the Golgi apparatus to the cell surface for recognition by CD8+ cytotoxic T lymphocytes (CTLs). In contrast, conventional MHC class II antigen presentation handles exogenous antigens, such as those from extracellular bacteria or soluble proteins, which are internalized by antigen-presenting cells (APCs) through endocytosis, phagocytosis, or receptor-mediated uptake. These antigens are delivered to endosomal-lysosomal compartments, where they are degraded by acid-dependent proteases, including cathepsins (e.g., cathepsin S), into peptides of 13-25 amino acids. MHC class II molecules, synthesized in the ER and protected by the invariant chain (Ii) to prevent premature peptide binding, traffic to specialized late endosomal compartments known as MHC class II compartments (MIICs). There, Ii is proteolyzed to leave the CLIP peptide, which is exchanged for antigenic peptides with the aid of HLA-DM; the resulting peptide-MHC II complexes are transported to the plasma membrane for presentation to CD4+ helper T cells. Cross-presentation fundamentally differs by enabling the presentation of exogenous antigens on molecules, thereby bridging the gap between extracellular threats and intracellular immune responses without requiring direct of the APC. Unlike conventional MHC I presentation, which relies on cytosolic antigens accessing the via , cross-presentation diverts endocytosed antigens away from the default MHC II pathway, often involving specialized mechanisms to access proteasomal degradation or endosomal loading while bypassing the need for antigens to enter the naturally. This process necessitates adaptations in certain APCs to prevent lysosomal degradation from routing antigens exclusively to MHC II, allowing for efficient peptide loading onto recycling or newly synthesized MHC I molecules. Consequently, cross-presentation permits CTL priming against pathogens or tumors that do not infect APCs, eliminating the requirement for physical contact between infected cells and APCs. By repurposing exogenous antigens for MHC I-restricted presentation, cross-presentation integrates the activation of + CTLs with + T cell help, which is typically driven by MHC II, thus linking humoral and cellular immunity in a coordinated response to extracellular antigens. This distinction underscores cross-presentation's critical role in initiating protective immunity against viruses, , and cancers that evade direct cytosolic entry, enhancing overall T cell-mediated surveillance.

Mechanisms of Cross-Presentation

Vacuolar Pathway

The vacuolar pathway of cross-presentation involves the processing of exogenous antigens entirely within endosomal or phagosomal compartments, without export to the . Antigens are internalized by antigen-presenting cells via or and degraded by lysosomal proteases such as cathepsin S and within these vacuoles. The resulting s are then loaded onto recycling or newly synthesized molecules in the same compartment, facilitating presentation to CD8+ T cells. This TAP-independent mechanism contrasts with the cytosolic pathway's reliance on for transport. Key molecular players regulate this process to optimize handling. Sec22b, an ER-Golgi intermediate compartment , promotes fusion between the and phagosomes, enabling delivery of loading machinery while controlling phagosomal maturation to prevent excessive acidification. , a phagosomal , generates that alkalinize the phagosome (to approximately 7.4), protecting antigens from rapid degradation by lysosomal enzymes and preserving integrity for MHC loading. This pathway offers advantages for handling soluble or particulate antigens, such as or immune complexes, due to its independence from proteasomal degradation and transport, allowing sustained presentation in certain contexts. However, it has limitations, including potential generation of peptides mismatched to those produced by direct cytosolic processing, which may reduce against certain tumor antigens compared to the cytosolic route. Experimental evidence supports the vacuolar pathway's distinct features, demonstrated in dendritic cells and macrophage cell lines. Treatment with , which blocks endosomal acidification and inhibits activity, significantly reduces cross-presentation via this route, confirming reliance on vacuolar . In contrast, lactacystin, a , does not impair vacuolar cross-presentation, underscoring its independence from cytosolic proteasomes.

Cytosolic Pathway

In the cytosolic pathway of cross-presentation, exogenous antigens internalized by or escape from or endosomes into the , where they undergo proteasomal degradation before peptide loading onto class I (MHC I) molecules. This translocation is facilitated by mechanisms such as endosomal membrane permeabilization and retrotranslocation machinery, allowing antigens to access the cytosolic proteasome for processing into short peptides (typically 8-10 ). The resulting peptides are then transported via the transporter associated with (TAP) into the (ER) or directly back into the phagosome, where they associate with nascent or recycled MHC I molecules for surface presentation to CD8+ T cells. Key molecular players in this pathway include components of the ER-associated degradation (ERAD) machinery, such as the AAA ATPase (VCP/p97), which drives antigen unfolding and extraction from endosomal membranes, while Derlin-1 does not appear essential. Autophagy-related protein 5 (ATG5) contributes to delivery by promoting formation, which facilitates the transport of endocytosed antigens to the for cross-presentation. Recent findings highlight perforin-2 (also known as ), a pore-forming protein expressed in dendritic cells (s), which permeabilizes endosomal membranes to enable , thereby supporting efficient cytosolic in -presenting cells. A 2025 study further identified WDFY4 as a key regulator in a shared pathway for cross-presentation of immune complexes, enhancing handling across subsets. This pathway is particularly advantageous for processing cell-associated antigens, such as those derived from apoptotic cells or infected tissues, due to its ability to generate peptides identical to those from endogenous sources, thereby ensuring consistent recognition by T cells. However, it is strictly dependent on functional and activity, rendering it sensitive to inhibitors like epoxomicin, which blocks proteasomal degradation and abolishes cross-presentation. Experimental evidence underscores the pathway's reliance on cytosolic processing, as demonstrated studies where TAP-deficient DCs exhibit severely impaired cross-priming of + T cells against exogenous . The ovalbumin (OVA) model , particularly tracking the SIINFEKL presentation on H-2Kb MHC I, has been instrumental in visualizing this , showing that cytosolic and proteasomal are required for efficient T cell in models.

Cross-Presenting Cells

Dendritic Cell Subsets

Dendritic cells (DCs) represent the principal antigen-presenting cells specialized for cross-presentation, with distinct subsets exhibiting varying efficiencies in capturing, processing, and presenting exogenous antigens to CD8⁺ T cells. Among conventional DCs, the cDC1 subset is uniquely equipped for robust cross-presentation, particularly of cell-associated antigens from viruses and tumors, enabling the priming of cytotoxic T lymphocyte responses. This specialization arises from their developmental dependence on the transcription factor Batf3, which is essential for their ontogeny and function in vivo. In mice, cDC1s are characterized by expression of CD8α, XCR1, and CLEC9A, while in humans, they are identified by XCR1 and CLEC9A (also known as BDCA3 or CD141). These cells preferentially employ the cytosolic pathway for antigen processing, supported by elevated expression of transporters associated with antigen processing (TAP) and enhanced endoplasmic reticulum-phagosome membrane contacts that facilitate antigen export to the cytosol for proteasomal degradation. Seminal studies in Batf3-deficient mice have demonstrated that the absence of cDC1s severely impairs cross-presentation and CD8⁺ T cell immunity against intracellular pathogens and tumors, underscoring their irreplaceable role. The cDC2 subset, in contrast, displays more limited proficiency in cross-presentation compared to cDC1s, primarily serving to prime CD4⁺ T cells through presentation. In mice, cDC2s express SIRPα and CD11b, whereas human counterparts are marked by CD1c and SIRPα (CD172a). Under inflammatory conditions, cDC2s can engage in cross-presentation via the vacuolar pathway, though with lower efficiency for cell-associated antigens, and they contribute modestly to ⁺ T cell activation in specific contexts such as certain infections. Their development relies on factors like , and they are more abundant in tissues, aiding in broader immune but with a focus on helper T cell responses rather than cytotoxic priming. Other DC populations, including plasmacytoid DCs (pDCs) and monocyte-derived DCs (moDCs), exhibit restricted cross-presentation capabilities. pDCs, identified by BDCA2 (CD303) and CD123 expression, primarily produce type I interferons and show minimal at steady state, with limited cross-presentation occurring via endosomal routes only upon or in the presence of adjuvants. Their phagocytic is low, particularly for large particles, limiting their role in this process. moDCs, generated from monocytes during inflammation and marked by and CD11b, demonstrate enhanced cross-presentation when stimulated by (TLR) ligands, such as those mimicking viral components, which upregulate machinery; however, they are transient and less efficient than conventional DCs . Skin-resident Langerhans cells (LCs), a specialized DC subset expressing CD207 (langerin) and residing in the , utilize endosomal pathways for cross-presentation of keratinocyte-derived , contributing to local immune that can foster tolerance in non-inflamed tissues. Across these subsets, functional adaptations optimize cross-presentation efficiency, including superior phagocytic uptake of apoptotic or necrotic material by cDC1s, chemotactic to lymph nodes via CCR7 expression, and rapid upregulation of costimulatory molecules like CD40 and to provide signals for full T cell activation. These features collectively enable DCs to bridge innate antigen capture with adaptive immunity, with cDC1s dominating in scenarios requiring strong CD8⁺ T cell responses.

Other Antigen-Presenting Cells

While dendritic cells are the primary professional antigen-presenting cells (APCs) specialized for efficient cross-presentation, other APCs such as macrophages and B cells also exhibit this capability, albeit with lower efficiency and in more context-dependent manners. Macrophages, particularly subsets like proinflammatory M1-like macrophages, can cross-present exogenous antigens via both vacuolar and cytosolic pathways, facilitating CD8+ T cell activation. These cells demonstrate high phagocytic uptake of apoptotic cells and debris, enabling antigen acquisition in tissues such as the spleen (red pulp macrophages), lymph nodes (subcapsular sinus macrophages), and tumors (CD206+ tumor-associated macrophages). However, macrophages often prioritize inflammatory responses over sustained T cell priming, with limited migration to lymph nodes compared to dendritic cells, restricting their role to local antigen presentation. Anti-inflammatory M2-like macrophages, in contrast, may promote tolerance through cross-presentation, as seen in Kupffer cells of the liver that induce CD8+ T cell apoptosis. B cells perform rare cross-presentation primarily of soluble antigens via the vacuolar route, which is enhanced by B cell receptor (BCR) crosslinking with immune complexes. This process supports limited + T cell priming, but B cells are more pivotal in and + T cell help rather than robust cytotoxic T lymphocyte (CTL) responses. Their cross-presentation efficiency is lower due to suboptimal costimulatory molecule expression (e.g., /), often leading to tolerogenic outcomes in autoreactive contexts like . Among other cells, neutrophils exhibit minimal cross-presentation under steady-state conditions but can be reprogrammed into antigen-presenting cells under inflammatory stress via Fcγ receptor engagement, enabling cross-presentation to CD8+ T cells with efficiency approaching that of dendritic cells in vitro. Epithelial cells, such as proximal tubular epithelial cells in the kidney, show limited capacity for cross-presentation, internalizing antigens via endocytosis and activating inflammatory CD8+ T cells in disease settings like lupus nephritis, though without strong costimulation. Overall, these non-dendritic APCs display reduced loading efficiency and costimulatory capacity relative to dendritic cells, frequently resulting in rather than effective priming, particularly in peripheral tissues.

Historical Development

Early Discoveries

The concept of cross-presentation began to emerge in the through studies on cytotoxic T lymphocyte (CTL) responses to minor (H) antigens, at a time when the principles of were still being established. In 1974, Zinkernagel and Doherty demonstrated that CTLs recognize viral antigens only when presented in the context of self-MHC molecules, establishing as a fundamental rule of T cell recognition. This framework highlighted that CTLs typically targeted antigens from infected or endogenous sources, yet observations of CTL activation against antigens from non-infecting cells suggested alternative pathways, particularly involving minor H antigens that differ between individuals despite shared MHC. A pivotal experiment in 1976 by provided the first clear evidence of what would later be termed cross-priming. Bevan injected mice with allogeneic cells expressing minor H antigens but differing in MHC (H-2) from the host; this induced CTL responses specific to the minor H antigens, restricted by the host's MHC, without the antigens being expressed directly in host cells. These CTLs could lyse target cells bearing the minor H antigens only when matched to the host's MHC, indicating that the priming occurred indirectly through host antigen-presenting cells capturing and presenting the exogenous antigens. This demonstration revealed that exogenous antigens from donor cells could access the pathway in recipient cells, challenging the emerging view that MHC class I presentation was limited to intracellularly synthesized proteins. Building on these findings, early evidence in the 1980s confirmed the phenomenon using bone marrow-derived antigen-presenting cells (APCs). In , , Carbone, and Bevan showed that bone marrow-derived macrophages could internalize and process exogenous soluble ovalbumin, presenting it on molecules to elicit responses from ovalbumin-specific, -restricted CTLs. This process required the macrophages to actively take up the soluble , distinguishing it from direct or endogenous expression, and underscored the role of specialized bone marrow-derived APCs in bridging exogenous antigens to the class I pathway. Collectively, these discoveries marked a conceptual shift, demonstrating that exogenous antigens could indeed enter the presentation pathway, thereby expanding the scope of CTL priming beyond infected cells.

Key Advances and Milestones

In the 1990s, seminal work from Ralph Steinman's laboratory and collaborators established dendritic cells (DCs) as the primary cross-presenting cells capable of processing exogenous antigens from apoptotic cells to stimulate MHC class I-restricted CD8+ T cell responses. A key study demonstrated that human DCs, but not macrophages, efficiently phagocytose apoptotic cell debris and present derived antigens on MHC class I molecules, inducing cytotoxic T lymphocytes (CTLs) in vitro. Concurrently, the discovery of the transporter associated with antigen processing (TAP), cloned in 1990, revealed its essential role in shuttling cytosolic peptides into the endoplasmic reticulum (ER) for MHC class I loading, which was soon extended to the cytosolic pathway of cross-presentation in DCs during the mid-to-late 1990s. These findings shifted the understanding of cross-presentation from a niche phenomenon to a DC-mediated process critical for initiating immunity against non-infectious threats like tumors. The 2000s brought advances in visualizing the cellular machinery underlying cross-presentation, particularly the ER-phagosome interactions that enable export to the . In 2003, researchers used and functional assays to show that phagosomes in DCs fuse with membranes, forming a hybrid compartment enriched in loading machinery, which supports TAP-dependent peptide transport and cross-presentation of phagocytosed antigens. By the end of the decade, genetic studies linked specific DC subsets to cross-priming efficiency; deletion of the Batf3 in mice ablated CD8α+ DCs, revealing their indispensable role in generating protective CD8+ T cell responses against viruses and tumors . The 2010s illuminated molecular transporters facilitating ER-phagosome communication and identified equivalents of murine cross-presenting DCs. In 2009–2011, investigations identified Sec22b, an ER-Golgi intermediate compartment , as a key mediator that pairs with plasma membrane SNAREs to promote phagosomal recruitment of ER components, thereby enhancing antigen crosspresentation without affecting phagosomal maturation. Additionally, characterization of BDCA3+ (CD141+) DCs expressing CLEC9A (DNGR-1) confirmed their equivalence to mouse CD8α+ DCs, with superior capacity for cross-presenting cell-associated antigens via CLEC9A-mediated uptake of necrotic debris. Recent 2020s developments have refined the mechanistic details and regulatory landscape of cross-presentation. A 2023 study elucidated the perforin-2 () mechanism in DCs, demonstrating that this pore-forming protein is recruited to phagosomal membranes, where it forms pores to enable antigen leakage into the for proteasomal degradation—a step essential for efficient crosspresentation and + T cell priming against tumors and pathogens. In 2025, analyses of platforms highlighted cross-presentation's dual role in priming and tolerance, showing that intramuscular mRNA delivery can induce cross-tolerance to self-antigens via DC-mediated presentation, informing strategies to mitigate in . Single-cell RNA sequencing (scRNA-seq) studies in the early further uncovered regulatory networks in cross-presenting DCs, revealing Irf8-dependent transcriptional clusters in pulmonary cDC1s that orchestrate genes for , ER-phagosome fusion, and T cell .

Roles in Immunity

Protective Responses Against Pathogens and Tumors

Cross-presentation plays a pivotal role in generating CD8+ T cell responses against intracellular pathogens by enabling dendritic cells (DCs) to present exogenous viral antigens on MHC class I molecules. In viral infections, such as those caused by lymphocytic choriomeningitis virus (LCMV) and influenza, antigens derived from infected apoptotic cells are captured by DCs, processed, and cross-presented to prime cytotoxic T lymphocytes (CTLs). This process is particularly critical for infections in non-hematopoietic tissues, where direct priming by infected professional antigen-presenting cells is limited, allowing surveillance and clearance of virus-infected cells. For instance, in LCMV infection, XCR1+ DCs cross-present viral antigens to CD8+ T cells, supporting their expansion and differentiation into effector cells essential for viral control. In the context of anti-tumor immunity, cross-presentation facilitates the activation of CTLs against tumor-associated antigens derived from dead or dying cancer cells. Conventional type 1 DCs (cDC1s) are the primary mediators, efficiently processing and presenting these antigens in tumor-draining lymph nodes to initiate + T cell responses. Within the , cDC1s further enhance CTL infiltration by producing such as CXCL9 and , which recruit CXCR3-expressing effector T cells, thereby promoting tumor clearance. This localized cross-presentation sustains anti-tumor effector functions, correlating with improved patient outcomes in various cancers. Mechanisms that enhance cross-presentation amplify these protective responses, broadening immunity through epitope spreading, where initial T cell activation against dominant epitopes leads to responses against subdominant or novel ones. Adjuvants like poly I:C, a TLR3 agonist, upregulate cross-presentation by promoting maturation and facilitating the acquisition of from apoptotic cells via receptors, resulting in stronger + T cell priming. This enhancement supports epitope spreading in both viral and tumor settings, enabling more comprehensive immune coverage against diverse antigens. Experimental models underscore the necessity of cross-presentation for protective immunity. In Batf3-/- mice, which lack cDC1s, + T cell priming against viral antigens is severely impaired during infections like and , leading to delayed viral clearance due to reduced effector responses in the lungs. Similarly, these mice exhibit defective anti-tumor CTL priming and fail to reject immunogenic tumors, highlighting the indispensable role of Batf3-dependent DCs in sustaining protective + T cell-mediated immunity.

Enhancement in Vaccination Strategies

Cross-presentation plays a pivotal role in enhancing efficacy by enabling the activation of cytotoxic + T cells (CTLs) against exogenous antigens, which is essential for combating intracellular pathogens and tumors. In vaccine design, strategies focus on directing antigens into cross-presentation pathways within dendritic cells (DCs) to promote robust CTL responses, often through the use of adjuvants and advanced delivery systems that mimic natural antigen uptake mechanisms. Subunit vaccines, which deliver purified antigens, frequently incorporate adjuvants that promote cross-presentation to overcome their inherent limitations in inducing + T cell immunity. Toll-like receptor (TLR) agonists, such as monophosphoryl lipid A (MPL) in the AS04 formulation, activate DCs to enhance uptake and processing via the cross-presentation pathway, leading to improved presentation. For instance, TLR2 agonists like SMIP 2.1 have been shown to stimulate cross-presentation in mouse bone marrow-derived DCs, resulting in stronger + T cell priming. Viral vector vaccines, particularly those based on adenovirus 5 (Ad5), mimic apoptotic delivery by infecting DCs or bystander cells, thereby facilitating endogenous and cross-presentation to elicit Th1-biased + T cell responses. These vectors have been instrumental in generating protective immunity in preclinical models by leveraging the cytosolic pathway for . Advanced delivery strategies further optimize cross-presentation by targeting specific cellular compartments. Nanoparticle encapsulation allows precise phagosomal targeting of antigens, where particle size influences endosomal escape and cross-presentation efficiency; for example, 20-200 nm nanoparticles promote antigen delivery to phagosomes in DCs, enhancing cytosolic translocation and MHC I loading when combined with TLR agonists. Electroporation enhances cytosolic export of DNA or mRNA vaccines by temporarily permeabilizing cell membranes, increasing antigen expression and access to the proteasome for cross-presentation, as demonstrated in studies where it boosted CD8+ T cell responses in murine models. Recent advancements in mRNA vaccines, as of 2025, utilize lipid nanoparticles (LNPs) optimized for cross-priming; for instance, mRNA-LNPs encoding tumor antigens are internalized by DCs to promote cross-presentation and antitumor CD8+ T cell responses through improved endosomal escape. These approaches yield improved CTL memory and response breadth, with cross-presentation being critical for success in several cases. Therapeutic HPV targeting E6 and E7 oncoproteins, such as mRNA-based platforms, rely on cross-presentation for generating + T cell responses against high-risk HPV types. Similarly, the rVSV-ZEBOV, a platform, induces cross-presentation-dependent + T cell responses that correlate with survival in nonhuman primates, highlighting the pathway's role in broad immunity against filoviruses. Despite these benefits, challenges persist in balancing effective priming with the risk of T cell exhaustion, particularly in targeting infections where prolonged exposure via cross-presentation can drive PD-1 upregulation and functional impairment. Strategies to mitigate exhaustion include combinations that limit excessive activation while sustaining memory CTL formation.

Roles in Tolerance

Peripheral Tolerance to Self-Antigens

Cross-presentation plays a crucial role in maintaining to self-antigens by enabling dendritic cells (DCs) to present exogenous self-antigens on molecules to + T cells, thereby inducing anergy, deletion, or regulatory responses that prevent . In the , without inflammatory cues, this process ensures that self-reactive T cells encountering their cognate antigens do not mount destructive responses. The primary mechanism involves the uptake of apoptotic or necrotic self-cells by DCs, particularly conventional DC1 (cDC1) subsets, which process and cross-present self-antigens via in a tolerogenic manner. Immature or steady-state DCs exhibit low levels of costimulatory molecules such as and , leading to incomplete T cell activation and promoting CD8+ T cell anergy or . Additionally, this low-costimulation environment favors the induction of regulatory T cells (Tregs), including + CD8+ Tregs, which suppress effector responses, or directly triggers cytotoxic T lymphocyte (CTL) deletion through Bim-dependent pathways. This tolerogenic cross-presentation occurs prominently in peripheral tissues such as the liver and during , where DCs continuously sample dying cells to avert responses against self-antigens released from physiological turnover. In the liver, resident DCs and sinusoidal endothelial cells cross-present antigens from apoptotic hepatocytes, contributing to the organ's inherent tolerogenic milieu. Similarly, in the , dermal DCs uptake antigens from apoptotic , enforcing to maintain barrier integrity without inflammation. Key regulators of this process include the absence of inflammatory signals, such as type I interferons (IFNs), which in steady-state conditions prevent DC maturation and proinflammatory production, thereby favoring over priming. The expression of programmed death-ligand 1 () on DCs further attenuates T cell activation by engaging PD-1 on self-reactive CD8+ T cells, promoting exhaustion or and reinforcing . Experimental evidence from mouse models demonstrates that cross-presentation of self-antigens, such as tissue-restricted antigens like ovalbumin expressed transgenically in pancreatic islets, induces CD8+ T cell tolerance through deletion or anergy when presented by CD8α+ DCs. Defects in cDC1 function, such as MHC class II deficiency, disrupt cross-tolerization, resulting in fatal autoimmunity driven by unchecked self-reactive CD8+ T cells.

Cross-Tolerance Mechanisms

Cross-tolerance mechanisms involve the cross-presentation of harmless exogenous antigens, such as those derived from dietary components or , by specialized dendritic cells (DCs) in the gut mucosa, resulting in the induction of regulatory T cells (Tregs) or anergy in + T cells to maintain peripheral immune . Gut-associated DCs, particularly CD103+ cDC1 subsets, acquire these antigens from the intestinal lumen or epithelial cells and process them for presentation on molecules to + T cells, promoting suppressive responses rather than effector immunity. This process extends tolerance mechanisms observed for self-antigens by specifically targeting environmental antigens to prevent aberrant . In the gut environment, dietary antigens like ovalbumin and commensal bacterial antigens are endocytosed by DCs, which migrate to mesenteric lymph nodes for cross-presentation, leading to IL-10 production by responding T cells and differentiation of + Tregs. For instance, CD103+ DCs imprint Tregs with gut-homing properties through (RA), enhancing IL-10 secretion and suppressive function against innocuous antigens. This IL-10-mediated pathway fosters Treg expansion, which in turn dampens pro-inflammatory + T cell activation, ensuring tolerance to persistent luminal challenges. At the molecular level, tolerogenic DCs preferentially utilize the vacuolar pathway for cross-presentation, where exogenous antigens are degraded in endolysosomal compartments by cathepsins, generating peptides that load onto recycling molecules without cytosolic escape, a process suited to low-inflammatory contexts. This pathway's dominance in tolerogenic DCs is complemented by and TGF-β signaling; , produced by CD103+ DCs via aldehyde dehydrogenase activity, synergizes with TGF-β to drive expression and Treg differentiation from naïve CD8+ or + T cells. TGF-β further modulates DC phenotype by upregulating inhibitory molecules like , reinforcing anergic states in antigen-specific T cells. Key contexts for cross-tolerance include oral tolerance to food antigens, where cross-presentation by intestinal DCs prevents allergic responses to dietary proteins, and microbiota-specific tolerance, which curbs chronic inflammation in the gut mucosa to avert conditions like (IBD). In oral tolerance, repeated exposure to food antigens via cross-presenting DCs induces peripheral Tregs that suppress Th2-driven . For commensals, cross-presentation maintains a balance where microbiota-derived antigens elicit IL-10-producing Tregs, protecting against dysbiosis-induced . Recent evidence from 2025 studies highlights PRDM16+ antigen-presenting cells (APCs), a RORγt-dependent of tolerogenic DCs, that present antigens to induce + Tregs, essential for oral and microbial . In Prdm16ΔRORγt mice, of these APCs abolishes Treg , resulting in elevated effector T cells, increased IgE, and allergic , demonstrating breakdown. Similarly, cDC1-deficient models show failed to epithelial antigens, leading to epithelial damage and Treg deficiency, underscoring the role in preventing IBD-like . These findings confirm that disrupting cross-presentation pathways precipitates loss of to exogenous gut antigens.

Clinical Implications

Applications in Cancer Immunotherapy

Cross-presentation by dendritic cells (DCs) plays a pivotal role in generating cytotoxic CD8+ T cell responses against tumor antigens, forming the basis for DC-based vaccines in . , the first FDA-approved autologous DC vaccine for metastatic castration-resistant , involves ex vivo loading of patient-derived antigen-presenting cells, including DCs, with a fusion protein of prostatic acid phosphatase and granulocyte-macrophage colony-stimulating factor, leading to a 22% reduction in the risk of death in phase III trials. Personalized neoantigen vaccines extend this approach by targeting patient-specific tumor mutations, with DCs pulsed or loaded ex vivo to promote cross-presentation of neoantigens via the cytosolic pathway, eliciting robust CD8+ T cell priming; clinical trials in and have shown induction of neoantigen-specific T cells correlating with tumor regression in up to 30% of patients. Tumor lysates, rich in intracellular antigens, are particularly effective for exploiting the cytosolic cross-presentation pathway in DCs, as they escape endosomal degradation and access the , enhancing CD8+ T cell responses in preclinical models and early-phase trials. Synergy between cross-presentation and inhibitors amplifies antitumor immunity, with PD-1 blockade enhancing the proliferation and effector function of cross-primed cytotoxic T lymphocytes (CTLs) by relieving exhaustion in the . In mouse models and human trials, PD-1 inhibition promotes spreading from cross-presented tumor antigens, broadening the T cell repertoire and improving response rates in immunogenic tumors like . As of 2025, ongoing clinical trials are exploring combinations of cDC1 expansion—using agents like Flt3L to mobilize Batf3-dependent cross-presenting DCs—with CAR-T therapies, aiming to boost and sustain CAR-T persistence; preliminary data from phase I studies indicate enhanced CTL infiltration and tumor control in solid tumors. Targeting strategies to augment cross-presentation include Batf3 agonists such as Flt3L, which mobilize and expand cross-presenting cDC1s in the tumor-draining nodes, potentiating + T cell priming and synergizing with checkpoint blockade in preclinical cancer models. Oncolytic viruses deliver tumor antigens directly to DCs by inducing immunogenic , facilitating cross-presentation and systemic antitumor immunity; for instance, engineered viruses expressing neoantigens have shown improved T cell responses and tumor regression in phase I/II trials for and . Clinical outcomes from cross-presentation-targeted therapies include improved survival in , where adjuvant vaccines loaded with tumor antigens have demonstrated immunological responses, such as antigen-specific T cell activation, though a phase III trial showed no significant extension in recurrence-free survival. However, challenges persist due to tumor-induced , which impairs function and cross-presentation efficiency, necessitating strategies like agonists to overcome these barriers and enhance therapeutic efficacy.

Strategies for Autoimmunity and Tolerance Induction

Therapeutic strategies leveraging cross-presentation aim to restore in autoimmune diseases by directing dendritic cells (DCs) to present autoantigens in a tolerogenic manner, thereby suppressing autoreactive T cell responses. vaccines represent a key approach, involving the encapsulation of autoantigens such as myelin basic protein (MBP) peptides within nanoparticles or liposomes to facilitate uptake and cross-presentation by tolerogenic DCs. This targeted delivery promotes the generation of regulatory T cells (Tregs) while minimizing inflammatory signaling, as demonstrated in preclinical models of (MS) where MBP peptide-loaded DCs induced antigen-specific tolerance and reduced disease severity. Rapamycin, an inhibitor, enhances these vaccines by biasing cross-presentation toward the vacuolar pathway in DCs, which favors tolerogenic processing over cytotoxic responses. In and animal studies show that rapamycin-treated DCs cross-present autoantigens like peptides more efficiently via endosomal compartments, leading to impaired + T cell activation and increased Treg induction, thereby attenuating autoimmune inflammation in models of and . Clinical translation includes phase I trials where vitamin D3-conditioned tolerogenic DCs loaded with MBP peptides were administered to MS patients, demonstrating safety and preliminary evidence of reduced autoreactive T cell proliferation. Engineering DCs for cross-tolerance further advances Treg induction by modifying cells to overexpress tolerogenic factors before reinfusion. For instance, DCs engineered to present autoantigens via cross-presentation pathways, combined with cytokines, have shown promise in inducing + Tregs that suppress effector T cells in autoimmune models. Recent 2025 research has identified PRDM16-dependent antigen-presenting cells crucial for promoting - and food antigen-specific peripheral Tregs in the gut, suggesting potential targets for therapies in and (IBD). Gene therapy strategies upregulate on cross-presenting DCs to reinforce by inhibiting T cell activation during . Viral vectors delivering genes to DCs have been shown in preclinical models to enhance to self-antigens, reducing autoreactive + T cell infiltration in tissues affected by . Nanoparticle-based delivery systems complement this by encapsulating autoantigens for targeted gut , where nanoparticles loaded with peptides are taken up by intestinal DCs for cross-presentation, inducing oral and Tregs in models of IBD and celiac disease. Evidence from phase I trials underscores the safety of these approaches in , with ongoing phase II studies evaluating efficacy of tolerogenic DCs for cross-presentation of antigens in dampening autoreactive responses while preserving protective immunity.

References

  1. [1]
    Current Concepts of Antigen Cross-Presentation - Frontiers
    The presentation of internalized antigens on MHC I molecules is a process termed cross-presentation. Efficient cross-presentation has been shown to be crucial ...
  2. [2]
    The biology and underlying mechanisms of cross-presentation of ...
    To monitor the health of cells, the immune system tasks antigen presenting cells with gathering antigens from other cells and reporting them to CD8 T cells ...
  3. [3]
    Regulation of the Cell Biology of Antigen Cross-Presentation
    Antigen cross-presentation is an adaptation of the cellular process of loading MHC-I molecules with endogenous peptides during their biosynthesis within the ...
  4. [4]
    Cross-presentation: underlying mechanisms and role in immune ...
    Cross-presentation is the only pathway by which the immune system can detect and respond to viral infections or mutations that exclusively occur in parenchymal ...Missing: definition | Show results with:definition
  5. [5]
    The MHC class I antigen presentation pathway: strategies for viral ...
    This review will discuss recent findings on some of the diverse array of mechanisms employed by human viruses to inhibit the MHC class I pathway in order to ...Missing: conventional seminal
  6. [6]
    The ins and outs of MHC class II-mediated antigen processing and ...
    In this Review, we describe our current knowledge of the mechanisms of uptake and processing of antigens, the intracellular formation of peptide–MHC class II ...
  7. [7]
    Current Concepts of Antigen Cross-Presentation - PMC - NIH
    The presentation of internalized antigens on MHC I molecules is a process termed cross-presentation. Efficient cross-presentation has been shown to be ...
  8. [8]
  9. [9]
    Pathways of MHC I cross-presentation of exogenous antigens - PMC
    There is a vacuolar pathway in which peptides are generated and then bind to MHC I molecules within the confines of the vacuole. Ingested exogenous antigens can ...
  10. [10]
  11. [11]
  12. [12]
  13. [13]
    Cross-presentation of exogenous antigens on MHC I molecules - PMC
    Specialized antigen presenting cells, such as DCs and macrophages, can present exogenous antigen on MHC-I in a process called cross-presentation.
  14. [14]
    Host immune system strikes back: Autophagy-mediated antigen ...
    Instead, antigen processing occurs within the vacuolar compartment and is blocked by inhibitors of lysosomal proteases such as chloroquine and leupeptin.
  15. [15]
    Regulation of Antigen Export to the Cytosol During Cross-Presentation
    Here we review recent advances on antigen cross-presentation, focusing on the mechanisms involved in antigen export to the cytosol, a crucial step of this ...
  16. [16]
    Cross-Presentation of Synthetic Long Peptides by Human Dendritic ...
    Our data support a role for the ER-associated degradation machinery (ERAD)-related protein p97/VCP in the transport of SLP16–40 from early endosomes to the ...
  17. [17]
    Perforin 2 mediates endosomal antigen export for cross-presentation
    May 3, 2023 · They define the pore-forming protein perforin 2 as an effector molecule that mediates antigen export from endosomes in cross-presenting cells.
  18. [18]
    Cross-Presentation of Cell-Associated Antigens by MHC ... - Frontiers
    In vivo, cross-presentation is biased toward highly expressed cellular antigens and favored by cellular destruction (18), including cellular destruction ...
  19. [19]
    Cellular protein is the source of cross-priming antigen in vivo - PNAS
    SIINFEKL-containing peptides are generated during the degradation of ovalbumin constructs in the cytosol by proteasomes. A fraction of these peptides are then ...
  20. [20]
    Dendritic cells in cancer immunology - Nature
    Sep 3, 2021 · This review covers three aspects of dendritic cell (DC) biology connected with antitumor immune responses but are not specifically oriented toward therapeutic ...
  21. [21]
    Batf3 Deficiency Reveals a Critical Role for CD8α+ Dendritic Cells in ...
    These results suggest an important role for CD8α + dendritic cells and cross-presentation in responses to viruses and in tumor rejection.
  22. [22]
    Human dendritic cell subsets: An updated view of their ontogeny ...
    Blood, skin, and lymphoid organ cDC1 are efficient cross‐presenting cells, with a superior ability for the cross‐presentation of necrotic cell‐associated ...
  23. [23]
    Dendritic cell subsets and implications for cancer immunotherapy
    1) cDC1s are responsible for priming CD8 T cells through B7/CD28 signaling, 2) cross-presentation of exogenous antigens on MHC class I molecules to CD8 T cells, ...Introduction · DC antigen presentation and T... · Dendritic cell subtypes · Conclusion
  24. [24]
    TLR ligands differentially affect uptake and presentation of cellular ...
    Jan 11, 2007 · Dendritic cells (DCs) have the unique ability to efficiently present T-cell epitopes from exogenous antigens on MHC class I molecules, ...
  25. [25]
    Antigen Cross-Presentation by Macrophages - Frontiers
    The aim of this review is to provide an overview of the current studies covering the roles and mechanisms of antigen cross-presentation by macrophages.Introduction · Cross-Presentation by Various... · Cellular Pathways of Cross...
  26. [26]
    Cross-presentation by the others - ScienceDirect.com
    Cross-presentation of cancer antigens has been described previously for CD206+ monocyte-derived cells13 upon CD47 blockade in bone marrow-derived ...Review · Introduction · Viral Antigen...<|control11|><|separator|>
  27. [27]
    CD206 + tumor-associated macrophages cross-present ... - JCI Insight
    May 3, 2022 · CD206+ macrophages cross-present tumor antigens, stimulating CD8+ T cells, and are associated with reduced tumor burden and improved survival.
  28. [28]
    A key role for cross-presentation of antibody-targeted antigens
    In this review, we summarize the published evidence from us and others, indicating that B cells can help T cells via cross-presentation of antibody-complexed ...
  29. [29]
    B-Cell Cross-Presentation of Autologous Antigen Precipitates ...
    B cells govern the transition from clinically silent insulitis to frank diabetes by cross-presenting autoantigen to self-reactive CD8 + T cells.B Cells Promote Cd8 Effector... · B-Cell Mhc Class I Is... · Role Of Cd4 T-Cell Help In...
  30. [30]
    FcγR engagement reprograms neutrophils into antigen cross ...
    Aug 9, 2021 · The latter process referred to as cross-presentation, is essential for anti-tumor immunity and eradication of pathogens. Among antigen- ...
  31. [31]
    Neutrophils efficiently cross-prime naive T cells in vivo | Blood
    We have shown that neutrophils cross-present ovalbumin to a CD8 + T-cell hybridoma and to naive CD8 + T cells from OT1 transgenic mice.Introduction · Patients, materials, and methods · Results · Discussion
  32. [32]
    Antigen Cross-Presentation by Murine Proximal Tubular Epithelial ...
    In this study, we aimed to investigate the capacity of PTECs for antigen cross-presentation thereby modulating CD8 + T-cell responses.
  33. [33]
    Langerhans cells cross-present antigen derived from skin - PNAS
    Cross-presentation is an important mechanism for generating immunity to viruses and tumors as well as for the induction of tolerance against self antigens (1, 2) ...
  34. [34]
    Cross-presenting Langerhans cells are required for the early ... - PNAS
    Aug 14, 2023 · Our data identify epidermal Langerhans cells that cross-present keratinocyte-derived antigens, as the professional APC indispensable for the early reactivation ...
  35. [35]
    Restriction of in vitro T cell-mediated cytotoxicity in lymphocytic ...
    Restriction of in vitro T cell-mediated cytotoxicity in ... Nature. 1974 Apr 19;248(5450):701-2. doi: 10.1038/248701a0. Authors. R M Zinkernagel, P C Doherty.
  36. [36]
    Cross-priming for a secondary cytotoxic response to minor H ...
    May 1, 1976 · Whether H-2 congenic lymphoid cells express minor histocompatibility determinants which cross-react at the cytotoxic T-cell level or the helper ...
  37. [37]
    ER–phagosome fusion defines an MHC class I cross ... - Nature
    Sep 25, 2003 · ER–phagosome fusion defines an MHC class I cross-presentation compartment in dendritic cells. Pierre Guermonprez,; Loredana Saveanu, ...
  38. [38]
    Batf3 deficiency reveals a critical role for CD8alpha+ dendritic cells ...
    Nov 14, 2008 · These results suggest an important role for CD8alpha+ dendritic cells and cross-presentation in responses to viruses and in tumor rejection.
  39. [39]
    Sec22b regulates phagosomal maturation and antigen ... - PubMed
    Dec 9, 2011 · Here, we identify the SNARE Sec22b as a specific regulator of Ag crosspresentation. Sec22b localizes to the ER-Golgi intermediate compartment ( ...Missing: 2009 paper
  40. [40]
    Characterization of human DNGR-1 + BDCA3 + leukocytes as ...
    May 17, 2010 · Here, we characterize a population of human DCs that expresses DNGR-1 (CLEC9A) and high levels of BDCA3 and resembles mouse CD8α+ DCs in ...
  41. [41]
    Perforin-2 is a pore-forming effector of endocytic escape in cross ...
    Jun 22, 2023 · Cross-presentation relies on the unusual “leakiness” of endocytic compartments in DCs, whereby internalized proteins escape into the cytosol for ...
  42. [42]
  43. [43]
    Single cell RNA sequencing reveals distinct clusters of Irf8 ...
    May 12, 2023 · Single cell RNA sequencing reveals distinct clusters of Irf8-expressing pulmonary conventional dendritic cells.Missing: networks | Show results with:networks
  44. [44]
    Apoptotic Cells Deliver Processed Antigen to Dendritic Cells for ...
    In sum, we demonstrated the in vivo relevance of DCs capturing processed antigen derived from dying cells for the cross-priming of CD8+ T cells, and suggest ...
  45. [45]
  46. [46]
  47. [47]
    The Role of Type 1 Conventional Dendritic Cells in Cancer Immunity
    Tumor-derived factors impair anti-tumor immunity by limiting cDC1 accumulation, survival, and function within the tumor microenvironment. DC Biology in Cancer.Dc Biology In Cancer · Box 1. Human Cdc1 · Priming Of T Cells After...
  48. [48]
  49. [49]
  50. [50]
    Essential role for Batf3-dependent dendritic cells in regulating CD8 ...
    Dec 27, 2023 · Upon challenge with respiratory influenza virus, Batf3-/- mice failed to induce protective immunity, suggesting a protective role for cDC1 in ...
  51. [51]
    Tumor-residing Batf3 dendritic cells are required for effector T ... - NIH
    Initial cross-priming of anti-tumor CD8+ T cells has been shown to depend upon Batf3-driven DC, and rejection of immunogenic tumors is ablated in Batf3−/− mice ...
  52. [52]
    Vaccine adjuvants to engage the cross-presentation pathway - PMC
    Aug 1, 2022 · In this review, we will focus on recent insights into molecular mechanisms of cross-presentation of exogenous antigens and the mechanism of ...Figure 2 · Antigen Transfer And Immune... · Adjuvants And...
  53. [53]
    Vaccine adjuvants: mechanisms and platforms - Nature
    Jul 19, 2023 · Adjuvants enhance the adaptive immunity of vaccines by activating innate immune cells. The core concept is that adjuvants promote the generation ...
  54. [54]
    A new TLR2 agonist promotes cross-presentation by mouse ... - NIH
    We investigated a new TLR2-based adjuvant, Small Molecule Immune Potentiator (SMIP) 2.1, for its ability to stimulate cross-presentation.
  55. [55]
    Adenovirus Vectors: Excellent Tools for Vaccine Development - PMC
    Adenovirus-based vaccines are currently being tested not only to prevent various infectious diseases but also to be applied as cancer vaccines.
  56. [56]
    Adenovirus vector and mRNA vaccines: Mechanisms regulating ...
    Nov 3, 2022 · The COVID-19 pandemic has seen the first widespread usage of two new vaccine technologies: replication-incompetent adenovirus (Ad) vectors and ...Abstract · Introduction · Adenovirus Vectors · mRNA vaccines
  57. [57]
    The Role of Nanovaccine in Cross-Presentation of Antigen ... - NIH
    Nov 15, 2019 · Collectively, nanoparticles incorporated with antigens and TLR agonists enhance the efficacy of vaccine, particularly for cellular immune ...
  58. [58]
    The role of phagosomal pH on the size-dependent efficiency of ...
    Vaccines able to stimulate CD8+ T cells are crucial in controlling a broad range of infectious diseases and tumors. To induce effective CD8+ T cell ...
  59. [59]
    Synthetic DNA Vaccines: Improved Vaccine Potency by ...
    Nov 4, 2013 · Two such improvements are the use of genetically engineered cytokine adjuvants and plasmid delivery via in vivo electroporation (EP), the latter ...
  60. [60]
    Dendritic cell targeting vaccine for HPV-associated cancer - PMC - NIH
    In line with this, we found that CD40 targeting leads to greater as well as prolonged antigen cross-presentation to CD8+ T cells compared to LOX-1 or Dectin-1 ...
  61. [61]
    Ebola virus vaccination elicits Ebola virus–specific immune ...
    Apr 2, 2025 · We showed that vaccine-induced responses were focused on the EBOV GP and did not cross-react substantially with other Ebolavirus proteins.
  62. [62]
    Vaccination Alters the Balance between Protective Immunity ...
    Our current study suggests that vaccines that tip the balance away from immune exhaustion and toward viral clearance may inadvertently increase transient ...
  63. [63]
    Exhausted T cells hijacking the cancer-immunity cycle - Frontiers
    One of the main drivers of T cell exhaustion is prolonged antigen presentation, a prerequisite in the cancer-immunity cycle. The umbrella term “T cell ...
  64. [64]
    The CD8α+ Dendritic Cell Is Responsible for Inducing Peripheral ...
    We previously described a mechanism for the maintenance of peripheral self-tolerance. This involves the cross-presentation of tissue-associated antigens by.Introduction · Materials And Methods · Results And Discussion
  65. [65]
    Self-antigen presentation by dendritic cells and lymphoid stroma ...
    In secondary lymphoid organs, dendritic cells (DCs) presenting self-antigen were thought to play a major role in the induction of peripheral T cell tolerance.
  66. [66]
    The Induction of Tolerance by Dendritic Cells That Have Captured ...
    We develop the hypothesis that immature DCs in the steady state are inducing tolerance to self-antigens within phagocytosed apoptotic bodies, derived from the ...
  67. [67]
    Dendritic cells as inducers of peripheral tolerance - PMC
    Although cross-presented antigens acquired from various tissues may lead to deletion of CD8+ T cells, the tolerance spontaneously induced by DCs in this way may ...
  68. [68]
    FoxP3+ regulatory T cells essentially contribute to peripheral ... - PNAS
    We show that antigen presentation by steady-state dendritic cells failed to induce peripheral tolerance in the absence of FoxP3 + regulatory T cells.<|control11|><|separator|>
  69. [69]
    Intestinal cDC1 drive cross-tolerance to epithelial-derived antigen ...
    Our results describe a role for FoxP3+CD8+ Tregs in cross-tolerance in the intestine for which development requires intestinal cDC1. INTRODUCTION. Peripheral ...Missing: costimulation | Show results with:costimulation
  70. [70]
    Antigen cross-presentation: extending recent laboratory findings to ...
    Also liver sinusoidal endothelial cells are capable of cross-presenting soluble exogenous antigen to CD8+ T cells leading to tolerance [16]. Other cell types ...
  71. [71]
    Self-Antigen Presentation by Dendritic Cells in ... - Frontiers
    The maintenance of peripheral tolerance requires self-antigen presentation by professional antigen presenting cells (APCs). Dendritic cells (DCs) are considered ...
  72. [72]
    Type I Interferons as Stimulators of DC-Mediated Cross-Priming
    Here, we will review the cross-presentation properties of different DC subsets, with special focus on cell-associated and tumor Ag, and discuss how IFN-I can ...
  73. [73]
    PD-L1 on dendritic cells attenuates T cell activation and regulates ...
    Sep 24, 2020 · PD-L1 on DCs is upregulated during antigen-presentation to protect DCs from cytotoxicity of activated T cells. However, such effect also dampens ...
  74. [74]
    Effective NY-ESO-1–specific MHC II–restricted T cell receptors ... - JCI
    Dec 10, 2018 · ... tolerance mechanisms implicated by NY-ESO-1 expression in the host. ... cross-presentation of NY-ESO-1 from tumor cells (Figure 7C and ...
  75. [75]
    Absence of MHC class II on cDC1 dendritic cells triggers fatal ...
    Mar 13, 2020 · The ability of cDC1s to enforce cross-tolerization is not completely acquired during homeostatic maturation but requires MHCII-restricted ...Absence Of Mhc Class Ii On... · Mhcii-Deficient Cdc1s Show... · Absence Of Mhcii Molecules...
  76. [76]
    Retinoic acid primes human dendritic cells to induce gut-homing, IL ...
    Jul 16, 2014 · Further analysis shows that the priming of IL-10-producing Treg cells is dependent on RA and is maintained in presence of inflammation. Thus, RA ...Missing: cross- | Show results with:cross-
  77. [77]
    Regulation of Intestinal Immune System by Dendritic Cells - PMC - NIH
    The presentation of innocuous food antigens by antigen-presenting cells induces oral tolerance by generation of inducible regulatory T (Treg) cells (14).
  78. [78]
    Intracellular Transport Routes for MHC I and Their Relevance for ...
    Early after the discovery of tolerogenic abilities of cross-presentation, it has ... In the proteasome-independent vacuolar pathway of cross-presentation ...Early Studies Of Mhc I... · Pathways Of Antigen... · Control Of Mhc I Antigen...<|control11|><|separator|>
  79. [79]
    Antigen-specific regulatory T-cell responses to intestinal microbiota
    Aug 2, 2017 · Failure to induce tolerance to commensal bacteria may underlie immune-mediated diseases such as human inflammatory bowel disease. At homeostasis ...
  80. [80]
    PRDM16-dependent antigen-presenting cells induce tolerance to ...
    Apr 14, 2025 · Here we identify an APC subset that is required for differentiation of both food- and microbiota-specific pT reg cells and for establishment of oral tolerance.
  81. [81]
    Sipuleucel-T Immunotherapy for Castration-Resistant Prostate Cancer
    Jul 29, 2010 · Sipuleucel-T, an autologous active cellular immunotherapy, has shown evidence of efficacy in reducing the risk of death among men with metastatic castration- ...
  82. [82]
    Neoantigen cancer vaccines: a new star on the horizon
    Apr 15, 2024 · Personalized neoantigen-based vaccines tailored to each patient have been developed that are capable of eliciting a robust and long-lasting immune response.Neoantigen-Targeted Adoptive... · Neoantigen-Based Cancer... · Neoantigen-Pulsed Dc...
  83. [83]
    Tumor cell lysates as immunogenic sources for cancer vaccine design
    The cross-presentation process has proved essential for the generation of cytotoxic T lymphocytes (CTLs) against viruses, transplanted cells, and tumor cells.
  84. [84]
    PD-1 blockade promotes epitope spreading in anticancer CD8+ T ...
    Our work constitutes the first report that interfering with PD-1 signaling potentiates epitope spreading in tumor-specific responses.
  85. [85]
    Engineered T cells stimulate dendritic cell recruitment and antigen ...
    Sep 16, 2025 · In Batf3−/− mice models, OT-I/XCL1-mediated tumor suppression was absent, emphasizing the essential role of endogenous cDC1s in the anti-tumor ...
  86. [86]
    Expanding cross-presenting dendritic cells enhances oncolytic ...
    Nov 22, 2022 · These data demonstrate that mobilizing DCs to increase tumor antigen cross-presentation improves oncolytic virotherapy.
  87. [87]
    Expression of tumor antigens within an oncolytic virus enhances the ...
    Jun 27, 2024 · However, through encoding tumor antigens within the virus, oncolytic virotherapy can generate anti-tumor T cell populations upon which immune ...Results · Discussion · Cell Lines And Viruses
  88. [88]
    Adjuvant dendritic cell therapy in stage IIIB/C melanoma - Nature
    Feb 23, 2024 · Autologous natural dendritic cells (nDCs) treatment can induce tumor-specific immune responses and clinical responses in cancer patients.
  89. [89]
    STING agonist-based ER-targeting molecules boost antigen cross ...
    Mar 26, 2025 · Cross-presentation of ABM5–OVA was substantially inhibited by MG132 but not by PD150606, indicating that the process follows a cytosolic pathway ...
  90. [90]
    Tolerogenic Dendritic Cells as a Promising Antigen-Specific ...
    A phase II trial using MBP83–99 was ... Antigen-specific tolerance by autologous myelin peptide-coupled cells: a phase 1 trial in multiple sclerosis.
  91. [91]
    Emerging Therapeutics for Immune Tolerance: Tolerogenic ...
    Other tolerogenic vaccine strategies included fusing myelin peptides to the immunosuppressive S-antigen from Plasmodium falciparum (128) or targeting myelin ...Abstract · Tolerogenic Vaccines for the... · Cell Therapy · Concluding Remarks
  92. [92]
    Use of Rapamycin in the Induction of Tolerogenic Dendritic Cells
    Rapamycin (RAPA), a macrocyclic triene antibiotic pro-drug, is a clinically-utilized 'tolerance-sparing' immunosuppressant that inhibits the activity of T, B, ...Missing: vacuolar pathway
  93. [93]
    Modulation of the mTOR pathway plays a central role in dendritic ...
    Aug 26, 2021 · The use of rapamycin decreased the expression of the cytokines tested, empowered the down-modulation of CD40 and also reduced splenocyte ...
  94. [94]
    Tolerogenic dendritic cell-based treatment for multiple sclerosis (MS)
    Sep 9, 2019 · A harmonised study protocol for two phase I clinical trials comparing intradermal and intranodal cell administrationMissing: cross- CTLs<|control11|><|separator|>
  95. [95]
    Tolerogenic Dendritic Cells and T-Regulatory Cells at the Clinical ...
    Tolerogenic dendritic cells and T-regulatory cells are two immune cell populations with the potential to prevent the onset of clinical stage type 1 diabetes.
  96. [96]
    Improved Immunological Tolerance Following Combination Therapy ...
    Sep 28, 2011 · For that, we aimed at stimulating the inhibitory PD-1 molecule expressed on T cells upon activation, by the gene transfer of its ligands PD-L1 ...
  97. [97]
    Nanoparticle-Based Drug Delivery Systems for Induction of ...
    Apr 6, 2022 · The application of nanoparticle-based drug delivery systems in autoimmune disease treatment can increase the efficiency of inducing antigen-specific tolerance ...
  98. [98]
    A Phase IIa Study of Vitamin D3 Tolerogenic Dendritic Cells (tolDC ...
    The investigators propose to design and conduct a phase IIa clinical trial to treat patients with multiple sclerosis (MS) by vaccination with tolerogenic ...Missing: II | Show results with:II
  99. [99]
    Antigen-specific immunotherapy via delivery of tolerogenic dendritic ...
    May 15, 2024 · Tolerogenic dendritic cells in MS were found to be safe in early clinical trials. Abstract. Multiple sclerosis (MS) is a chronic inflammatory ...