Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.862
Filter
1.
Front Immunol ; 15: 1411490, 2024.
Article in English | MEDLINE | ID: mdl-39139570

ABSTRACT

Immunotherapy has revolutionized cancer treatment by leveraging the immune system's innate capabilities to combat malignancies. Despite the promise of tumor antigens in stimulating anti-tumor immune responses, their clinical utility is hampered by limitations in eliciting robust and durable immune reactions, exacerbated by tumor heterogeneity and immune evasion mechanisms. Recent insights into the immunogenic properties of host homologous microbial antigens have sparked interest in their potential for augmenting anti-tumor immunity while minimizing off-target effects. This review explores the therapeutic potential of microbial antigen peptides in tumor immunotherapy, beginning with an overview of tumor antigens and their challenges in clinical translation. We further explore the intricate relationship between microorganisms and tumor development, elucidating the concept of molecular mimicry and its implications for immune recognition of tumor-associated antigens. Finally, we discuss methodologies for identifying and characterizing microbial antigen peptides, highlighting their immunogenicity and prospects for therapeutic application.


Subject(s)
Antigens, Bacterial , Antigens, Neoplasm , Immunotherapy , Neoplasms , Humans , Antigens, Neoplasm/immunology , Neoplasms/immunology , Neoplasms/therapy , Immunotherapy/methods , Animals , Antigens, Bacterial/immunology , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Molecular Mimicry/immunology
2.
Nature ; 632(8025): 622-629, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39112696

ABSTRACT

Multisystem inflammatory syndrome in children (MIS-C) is a severe, post-infectious sequela of SARS-CoV-2 infection1,2, yet the pathophysiological mechanism connecting the infection to the broad inflammatory syndrome remains unknown. Here we leveraged a large set of samples from patients with MIS-C to identify a distinct set of host proteins targeted by patient autoantibodies including a particular autoreactive epitope within SNX8, a protein involved in regulating an antiviral pathway associated with MIS-C pathogenesis. In parallel, we also probed antibody responses from patients with MIS-C to the complete SARS-CoV-2 proteome and found enriched reactivity against a distinct domain of the SARS-CoV-2 nucleocapsid protein. The immunogenic regions of the viral nucleocapsid and host SNX8 proteins bear remarkable sequence similarity. Consequently, we found that many children with anti-SNX8 autoantibodies also have cross-reactive T cells engaging both the SNX8 and the SARS-CoV-2 nucleocapsid protein epitopes. Together, these findings suggest that patients with MIS-C develop a characteristic immune response to the SARS-CoV-2 nucleocapsid protein that is associated with cross-reactivity to the self-protein SNX8, demonstrating a mechanistic link between the infection and the inflammatory syndrome, with implications for better understanding a range of post-infectious autoinflammatory diseases.


Subject(s)
Antibodies, Viral , Autoantibodies , COVID-19 , Cross Reactions , Epitopes , Molecular Mimicry , SARS-CoV-2 , Systemic Inflammatory Response Syndrome , Child , Humans , Antibodies, Viral/immunology , Autoantibodies/immunology , Coronavirus Nucleocapsid Proteins/chemistry , Coronavirus Nucleocapsid Proteins/immunology , COVID-19/immunology , COVID-19/virology , COVID-19/complications , Cross Reactions/immunology , Epitopes/immunology , Epitopes/chemistry , Molecular Mimicry/immunology , Phosphoproteins/chemistry , Phosphoproteins/immunology , SARS-CoV-2/chemistry , SARS-CoV-2/immunology , SARS-CoV-2/metabolism , SARS-CoV-2/pathogenicity , Sorting Nexins/chemistry , Sorting Nexins/immunology , Systemic Inflammatory Response Syndrome/immunology , Systemic Inflammatory Response Syndrome/pathology , Systemic Inflammatory Response Syndrome/virology , T-Lymphocytes/immunology
3.
Biomolecules ; 14(8)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39199321

ABSTRACT

Epidemiological studies and meta-analyses have shown a strong association between high seroprevalence of Toxoplasma gondii (T. gondii) and schizophrenia. Schizophrenic patients showed higher levels of anti-Toxoplasma immunoglobulins M and G (IgM and IgG) when compared to healthy controls. Previously, in a rat model, we demonstrated that the progeny of mothers immunized with T. gondii lysates before gestation had behavioral and social impairments during adulthood. Therefore, we suggested that T. gondii infection can trigger autoreactivity by molecularly mimicking host brain proteins. Here, we aimed to identify the occurrence of antigenic mimicry between T. gondii epitopes and host brain proteins. Using a bioinformatic approach, we predicted T. gondii RH-88 B cell epitopes and compared them to human cell-surface proteins involved in brain development and differentiation (BrainS). Five different algorithms for B-cell-epitope prediction were used and compared, resulting in 8584 T. gondii epitopes. We then compared T. gondii predicted epitopes to BrainS proteins by local sequence alignments using BLASTP. T. gondii immunogenic epitopes significantly overlapped with 42 BrainS proteins. Among these overlapping proteins essential for brain development and differentiation, we identified HSP90 and NOTCH receptors as the proteins most likely to be targeted by the maternally generated pathogenic antibodies due to their topological overlap at the extracellular region of their sequence. This analysis highlights the relevance of pregestational clinical surveillance and screening for potential pathogenic anti-T. gondii antibodies. It also identifies potential targets for the design of vaccines that could prevent behavioral and cognitive impairments associated with pre-gestational T. gondii exposure.


Subject(s)
Brain , Epitopes, B-Lymphocyte , Molecular Mimicry , Toxoplasma , Toxoplasma/immunology , Molecular Mimicry/immunology , Humans , Epitopes, B-Lymphocyte/immunology , Brain/parasitology , Brain/immunology , Brain/metabolism , Computational Biology/methods , Toxoplasmosis/immunology , Animals , Antibodies, Protozoan/immunology , Rats
4.
J Biomed Sci ; 31(1): 83, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39169357

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) vaccine immunogens capable of inducing broadly neutralizing antibodies (bNAbs) remain obscure. HIV-1 evades immune responses through enormous diversity and hides its conserved vulnerable epitopes on the envelope glycoprotein (Env) by displaying an extensive immunodominant glycan shield. In elite HIV-1 viremic controllers, glycan-dependent bNAbs targeting conserved Env epitopes have been isolated and are utilized as vaccine design templates. However, immunological tolerance mechanisms limit the development of these antibodies in the general population. The well characterized bNAbs monoclonal variants frequently exhibit extensive levels of somatic hypermutation, a long third heavy chain complementary determining region, or a short third light chain complementarity determining region, and some exhibit poly-reactivity to autoantigens. This review elaborates on the obstacles to engaging and manipulating the Env glycoprotein as an effective immunogen and describes an alternative reverse vaccinology approach to develop a novel category of bNAb-epitope-derived non-cognate immunogens for HIV-1 vaccine design.


Subject(s)
AIDS Vaccines , Antibodies, Neutralizing , HIV Antibodies , HIV-1 , HIV-1/immunology , Humans , AIDS Vaccines/immunology , Antibodies, Neutralizing/immunology , HIV Antibodies/immunology , Polysaccharides/immunology , HIV Infections/immunology , Molecular Mimicry/immunology , Epitopes/immunology , env Gene Products, Human Immunodeficiency Virus/immunology , Ligands
5.
Nat Commun ; 15(1): 6408, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39164229

ABSTRACT

Venomous animals have evolved diverse molecular mechanisms to incapacitate prey and defend against predators. Most venom components disrupt nervous, locomotor, and cardiovascular systems or cause tissue damage. The discovery that certain fish-hunting cone snails use weaponized insulins to induce hypoglycemic shock in prey highlights a unique example of toxins targeting glucose homeostasis. Here, we show that, in addition to insulins, the deadly fish hunter, Conus geographus, uses a selective somatostatin receptor 2 (SSTR2) agonist that blocks the release of the insulin-counteracting hormone glucagon, thereby exacerbating insulin-induced hypoglycemia in prey. The native toxin, Consomatin nG1, exists in several proteoforms with a minimized vertebrate somatostatin-like core motif connected to a heavily glycosylated N-terminal region. We demonstrate that the toxin's N-terminal tail closely mimics a glycosylated somatostatin from fish pancreas and is crucial for activating the fish SSTR2. Collectively, these findings provide a stunning example of chemical mimicry, highlight the combinatorial nature of venom components, and establish glucose homeostasis as an effective target for prey capture.


Subject(s)
Conus Snail , Glucagon , Glucose , Homeostasis , Insulin , Receptors, Somatostatin , Somatostatin , Animals , Somatostatin/metabolism , Homeostasis/drug effects , Insulin/metabolism , Glucose/metabolism , Receptors, Somatostatin/metabolism , Glucagon/metabolism , Fishes/metabolism , Predatory Behavior/drug effects , Hypoglycemia/metabolism , Mollusk Venoms/metabolism , Humans , Molecular Mimicry
6.
Nat Commun ; 15(1): 7294, 2024 Aug 24.
Article in English | MEDLINE | ID: mdl-39181890

ABSTRACT

To enter epithelial cells, the obligate intracellular pathogen Chlamydia pneumoniae secretes early effector proteins, which bind to and modulate the host-cell's plasma membrane and recruit several pivotal endocytic host proteins. Here, we present the high-resolution structure of an entry-related chlamydial effector protein, SemD. Co-crystallisation of SemD with its host binding partners demonstrates that SemD co-opts the Cdc42 binding site to activate the actin cytoskeleton regulator N-WASP, making active, GTP-bound Cdc42 superfluous. While SemD binds N-WASP much more strongly than Cdc42 does, it does not bind the Cdc42 effector protein FMNL2, indicating effector protein specificity. Furthermore, by identifying flexible and structured domains, we show that SemD can simultaneously interact with the membrane, the endocytic protein SNX9, and N-WASP. Here, we show at the structural level how a single effector protein can hijack central components of the host's endocytic system for efficient internalization.


Subject(s)
Bacterial Proteins , Chlamydophila pneumoniae , Endocytosis , Wiskott-Aldrich Syndrome Protein, Neuronal , cdc42 GTP-Binding Protein , Humans , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Binding Sites , cdc42 GTP-Binding Protein/metabolism , Cell Membrane/metabolism , Chlamydophila pneumoniae/metabolism , Crystallography, X-Ray , HeLa Cells , Host-Pathogen Interactions , Molecular Mimicry , Protein Binding , Sorting Nexins/metabolism , Sorting Nexins/chemistry , Sorting Nexins/genetics , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism , Animals , Rats
7.
J Immunotoxicol ; 21(1): 2373247, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39066679

ABSTRACT

Molecular mimicry has been proposed to be a possible mechanism of induction of autoimmunity. In some cases, it is believed that such events could lead to a disease such as Type 1 diabetes (T1D). One of the primary MHC-I epitopes in the non-obese diabetic (NOD) mouse model of T1D has been identified as a peptide from the islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP) protein. In humans, the most common MHC-I model allele is HLA-A02; based on this, the study here identified a potential HLA-A0201-restricted human IGRP epitope as YLKTNLFLFL and also found a homologous A0201-restricted peptide in an Enterococcal protein. Using cells obtained from healthy human donors, it was seen that after a 2-week incubation with the synthetic bacterial protein, healthy A0201+ donor CD8+ cells displayed increased staining for human IGRP-peptide-dextramer. On the other hand, in control cultures, no significant levels of dextramer-staining CD8+ T-cells were detectable. From these outcomes, it is possible to conclude that certain bacterial proteins may initiate CD8+ T-cell-mediated immune reaction toward homologous human antigens.


Subject(s)
Antigens, Bacterial , CD8-Positive T-Lymphocytes , Cross Reactions , Diabetes Mellitus, Type 1 , Epitopes, T-Lymphocyte , Glucose-6-Phosphatase , HLA-A2 Antigen , Humans , Diabetes Mellitus, Type 1/immunology , HLA-A2 Antigen/immunology , HLA-A2 Antigen/metabolism , Antigens, Bacterial/immunology , Glucose-6-Phosphatase/immunology , Glucose-6-Phosphatase/genetics , Cross Reactions/immunology , Epitopes, T-Lymphocyte/immunology , CD8-Positive T-Lymphocytes/immunology , Animals , Mice , Molecular Mimicry/immunology , Mice, Inbred NOD , Bacterial Proteins/immunology , Cells, Cultured
8.
Adv Protein Chem Struct Biol ; 142: 421-436, 2024.
Article in English | MEDLINE | ID: mdl-39059993

ABSTRACT

Host-pathogen interactions are complex associations which evolve over long co-evolutionary histories. Pathogens exhibit different mechanisms to gain advantage over their host. Mimicry of host factors is an influential tool in subverting host mechanisms to ensure pathogenesis. This chapter discusses such molecular mimicry exhibited during viral infections. Understanding the evolutionary relationships, shared identity and functional impact of the virus encoded mimics is critical. With a particular emphasis on viral mimics and their association with cancer and autoimmune diseases, this chapter highlights the importance of molecular mimicry in virus biology.


Subject(s)
Molecular Mimicry , Humans , Viruses/metabolism , Host-Pathogen Interactions , Virus Diseases/metabolism , Virus Diseases/virology , Virus Diseases/immunology , Endocrine System/metabolism , Neoplasms/metabolism , Neoplasms/virology , Animals , Autoimmune Diseases/metabolism , Autoimmune Diseases/virology , Autoimmune Diseases/immunology
9.
Biochemistry ; 63(15): 1955-1962, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39012171

ABSTRACT

In mammals, cobalamin is an essential cofactor that is delivered by a multitude of chaperones in an elaborate trafficking pathway to two client enzymes, methionine synthase and methylmalonyl-CoA mutase (MMUT). Rhodibalamins, the rhodium analogs of cobalamins, have been described as antimetabolites due to their ability to inhibit bacterial growth. In this study, we have examined the reactivity of adenosylrhodibalamin (AdoRhbl) with two key human chaperones, MMACHC (also known as CblC) and adenosyltransferase (MMAB, also known as ATR), and with the human and Mycobacterium tuberculosis MMUT. We demonstrate that while AdoRhbl binds tightly to all four proteins, the Rh-carbon bond is resistant to homolytic (on MMAB and MMUT) as well as heterolytic (on MMACHC) rupture. On the other hand, MMAB catalyzes Rh-carbon bond formation, converting rhodi(I)balamin in the presence of ATP to AdoRhbl. We report the first crystal structure of a rhodibalamin (AdoRhbl) bound to a B12 protein, i.e., MMAB, in the presence of triphosphate, which shows a weakened but intact Rh-carbon bond. The structure provides insights into how MMAB cleaves the corresponding Co-carbon bond in a sacrificial homolytic reaction that purportedly functions as a cofactor sequestration strategy. Collectively, the study demonstrates that while the noble metal substitution of cobalt by rhodium sets up structural mimicry, it compromises chemistry, which could be exploited for targeting human and bacterial B12 chaperones and enzymes.


Subject(s)
Vitamin B 12 , Vitamin B 12/metabolism , Vitamin B 12/chemistry , Vitamin B 12/analogs & derivatives , Humans , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/metabolism , Methylmalonyl-CoA Mutase/metabolism , Methylmalonyl-CoA Mutase/chemistry , Rhodium/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Molecular Chaperones/metabolism , Molecular Chaperones/chemistry , Molecular Mimicry , Models, Molecular , Alkyl and Aryl Transferases
10.
Scand J Immunol ; 99(2): e13344, 2024 Feb.
Article in English | MEDLINE | ID: mdl-39007954

ABSTRACT

COVID-19 is a severe infectious disease caused by a SARS-CoV-2 infection. It has caused a global pandemic and can lead to acute respiratory distress syndrome (ARDS). Beyond the respiratory system, the disease manifests in multiple organs, producing a spectrum of clinical symptoms. A pivotal factor in the disease's progression is autoimmunity, which intensifies its severity and contributes to multi-organ injuries. The intricate interaction between the virus' spike protein and human proteins may engender the generation of autoreactive antibodies through molecular mimicry. This can further convolute the immune response, with the potential to escalate into overt autoimmunity. There is also emerging evidence to suggest that COVID-19 vaccinations might elicit analogous autoimmune responses. Advanced technologies have pinpointed self-reactive antibodies that target diverse organs or immune-modulatory proteins. The interplay between autoantibody levels and multi-organ manifestations underscores the importance of regular monitoring of serum antibodies and proinflammatory markers. A combination of immunosuppressive treatments and antiviral therapy is crucial for managing COVID-19-associated autoimmune diseases. The review will focus on the generation of autoantibodies in the context of COVID-19 and their impact on organ health.


Subject(s)
Autoantibodies , Autoimmunity , COVID-19 , SARS-CoV-2 , Humans , COVID-19/immunology , Autoantibodies/immunology , SARS-CoV-2/immunology , Autoimmunity/immunology , Autoimmune Diseases/immunology , Molecular Mimicry/immunology , Spike Glycoprotein, Coronavirus/immunology , Antibodies, Viral/immunology
12.
J Am Chem Soc ; 146(31): 22093-22102, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39054926

ABSTRACT

Here we introduce amphiphilic star polymers as versatile protein mimics capable of approximating the activity of certain native proteins. Our study focuses on designing a synthetic polymer capable of replicating the biological activity of TRAIL, a promising anticancer protein that shows very poor circulation half-life. Successful protein mimicry requires precise control over the presentation of receptor-binding peptides from the periphery of the polymer scaffold while maintaining enough flexibility for protein-peptide binding. We show that this can be achieved by building hydrophobic blocks into the core of a star-shaped polymer, which drives unimolecular collapse in water. By screening a library of diblock copolymer stars, we were able to design structures with IC50's of ∼4 nM against a colon cancer cell line (COLO205), closely approximating the activity of the native TRAIL protein. This finding highlights the broad potential for simple synthetic polymers to mimic the biological activity of complex proteins.


Subject(s)
Polymers , Humans , Polymers/chemistry , Polymers/pharmacology , Cell Line, Tumor , TNF-Related Apoptosis-Inducing Ligand/chemistry , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Hydrophobic and Hydrophilic Interactions , Molecular Mimicry , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology
13.
Front Immunol ; 15: 1398002, 2024.
Article in English | MEDLINE | ID: mdl-38947322

ABSTRACT

Background: In the present study we investigated whether peptides derived from the entire SARS-CoV-2 proteome share homology to TAAs (tumor-associated antigens) and cross-reactive CD8+ T cell can be elicited by the BNT162b2 preventive vaccine or the SARS-CoV-2 natural infection. Methods and results: Viral epitopes with high affinity (<100nM) to the HLA-A*02:01 allele were predicted. Shared and variant-specific epitopes were identified. Significant homologies in amino acidic sequence have been found between SARS-CoV-2 peptides and multiple TAAs, mainly associated with breast, liver, melanoma and colon cancers. The molecular mimicry of the viral epitopes and the TAAs was found in all viral proteins, mostly the Orf 1ab and the Spike, which is included in the BNT162b2 vaccine. Predicted structural similarities confirmed the sequence homology and comparable patterns of contact with both HLA and TCR α and ß chains were observed. CD8+ T cell clones cross-reactive with the paired peptides have been found by MHC class l-dextramer staining. Conclusions: Our results show for the first time that several SARS-COV-2 antigens are highly homologous to TAAs and cross-reactive T cells are identified in infected and BNT162b2 preventive vaccinated individuals. The implication would be that the SARS-Cov-2 pandemic could represent a natural preventive immunization for breast, liver, melanoma and colon cancers. In the coming years, real-world evidences will provide the final proof for such immunological experimental evidence. Moreover, such SARS-CoV-2 epitopes can be used to develop "multi-cancer" off-the-shelf preventive/therapeutic vaccine formulations, with higher antigenicity and immunogenicity than over-expressed tumor self-antigens, for the potential valuable benefit of thousands of cancer patients around the World.


Subject(s)
CD8-Positive T-Lymphocytes , COVID-19 , Cross Reactions , Epitopes, T-Lymphocyte , Molecular Mimicry , SARS-CoV-2 , Humans , SARS-CoV-2/immunology , COVID-19/prevention & control , COVID-19/immunology , Molecular Mimicry/immunology , CD8-Positive T-Lymphocytes/immunology , Cross Reactions/immunology , Epitopes, T-Lymphocyte/immunology , BNT162 Vaccine/immunology , Antigens, Viral/immunology , HLA-A2 Antigen/immunology , Neoplasms/immunology , Neoplasms/prevention & control , Antigens, Neoplasm/immunology , COVID-19 Vaccines/immunology
14.
Eur J Clin Microbiol Infect Dis ; 43(8): 1505-1516, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38829448

ABSTRACT

The relationship between infectious agents and autoimmune diseases is a complex issue. In recent years, increasing clinical cases have indicated that infectious agents play an important role in the development of autoimmune diseases. Molecular mimicry is currently widely regarded as the primary pathogenic mechanism of various autoimmune diseases in humans. Components of infectious agents can undergo molecular mimicry with components in patients' bodies, leading to the development of various autoimmune diseases. In this article, we provide a brief overview of current research of the current research status on the relationship between infectious agents and autoimmune diseases, and describe our current understanding of their mechanisms of action in order to better understand the pathogenesis, diagnosis, and treatment of autoimmune diseases.


Subject(s)
Autoimmune Diseases , Humans , Molecular Mimicry , Communicable Diseases
15.
PLoS Pathog ; 20(6): e1012300, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38900818

ABSTRACT

The AAA-type ATPase VPS4 is recruited by proteins of the endosomal sorting complex required for transport III (ESCRT-III) to catalyse membrane constriction and membrane fission. VPS4A accumulates at the cytoplasmic viral assembly complex (cVAC) of cells infected with human cytomegalovirus (HCMV), the site where nascent virus particles obtain their membrane envelope. Here we show that VPS4A is recruited to the cVAC via interaction with pUL71. Sequence analysis, deep-learning structure prediction, molecular dynamics and mutagenic analysis identify a short peptide motif in the C-terminal region of pUL71 that is necessary and sufficient for the interaction with VPS4A. This motif is predicted to bind the same groove of the N-terminal VPS4A Microtubule-Interacting and Trafficking (MIT) domain as the Type 2 MIT-Interacting Motif (MIM2) of cellular ESCRT-III components, and this viral MIM2-like motif (vMIM2) is conserved across ß-herpesvirus pUL71 homologues. However, recruitment of VPS4A by pUL71 is dispensable for HCMV morphogenesis or replication and the function of the conserved vMIM2 during infection remains enigmatic. VPS4-recruitment via a vMIM2 represents a previously unknown mechanism of molecular mimicry in viruses, extending previous observations that herpesviruses encode proteins with structural and functional homology to cellular ESCRT-III components.


Subject(s)
Cytomegalovirus , Endosomal Sorting Complexes Required for Transport , Molecular Mimicry , Vacuolar Proton-Translocating ATPases , Virus Assembly , Humans , Endosomal Sorting Complexes Required for Transport/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Vacuolar Proton-Translocating ATPases/genetics , Cytomegalovirus/metabolism , Cytomegalovirus/genetics , Cytomegalovirus/physiology , Virus Assembly/physiology , Cytomegalovirus Infections/virology , Cytomegalovirus Infections/metabolism , ATPases Associated with Diverse Cellular Activities/metabolism , ATPases Associated with Diverse Cellular Activities/genetics , Viral Proteins/metabolism , Viral Proteins/genetics
17.
Int J Mol Sci ; 25(11)2024 May 21.
Article in English | MEDLINE | ID: mdl-38891798

ABSTRACT

Systemic lupus erythematosus (SLE) is a multifactorial autoimmune disease characterized by self-immune tolerance breakdown and the production of autoantibodies, causing the deposition of immune complexes and triggering inflammation and immune-mediated damage. SLE pathogenesis involves genetic predisposition and a combination of environmental factors. Clinical manifestations are variable, making an early diagnosis challenging. Heat shock proteins (Hsps), belonging to the chaperone system, interact with the immune system, acting as pro-inflammatory factors, autoantigens, as well as immune tolerance promoters. Increased levels of some Hsps and the production of autoantibodies against them are correlated with SLE onset and progression. The production of these autoantibodies has been attributed to molecular mimicry, occurring upon viral and bacterial infections, since they are evolutionary highly conserved. Gut microbiota dysbiosis has been associated with the occurrence and severity of SLE. Numerous findings suggest that proteins and metabolites of commensal bacteria can mimic autoantigens, inducing autoimmunity, because of molecular mimicry. Here, we propose that shared epitopes between human Hsps and those of gut commensal bacteria cause the production of anti-Hsp autoantibodies that cross-react with human molecules, contributing to SLE pathogenesis. Thus, the involvement of the chaperone system, gut microbiota dysbiosis, and molecular mimicry in SLE ought to be coordinately studied.


Subject(s)
Dysbiosis , Gastrointestinal Microbiome , Lupus Erythematosus, Systemic , Molecular Mimicry , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/microbiology , Lupus Erythematosus, Systemic/metabolism , Humans , Molecular Mimicry/immunology , Dysbiosis/immunology , Gastrointestinal Microbiome/immunology , Molecular Chaperones/metabolism , Molecular Chaperones/immunology , Heat-Shock Proteins/immunology , Heat-Shock Proteins/metabolism , Autoantibodies/immunology , Animals , Autoantigens/immunology , Autoantigens/metabolism , Autoimmunity
18.
Proc Natl Acad Sci U S A ; 121(24): e2316892121, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38833472

ABSTRACT

The loss of function of AAA (ATPases associated with diverse cellular activities) mechanoenzymes has been linked to diseases, and small molecules that activate these proteins can be powerful tools to probe mechanisms and test therapeutic hypotheses. Unlike chemical inhibitors that can bind a single conformational state to block enzyme function, activator binding must be permissive to different conformational states needed for mechanochemistry. However, we do not know how AAA proteins can be activated by small molecules. Here, we focus on valosin-containing protein (VCP)/p97, an AAA unfoldase whose loss of function has been linked to protein aggregation-based disorders, to identify druggable sites for chemical activators. We identified VCP ATPase Activator 1 (VAA1), a compound that dose-dependently stimulates VCP ATPase activity up to ~threefold. Our cryo-EM studies resulted in structures (ranging from ~2.9 to 3.7 Å-resolution) of VCP in apo and ADP-bound states and revealed that VAA1 binds an allosteric pocket near the C-terminus in both states. Engineered mutations in the VAA1-binding site confer resistance to VAA1, and furthermore, modulate VCP activity. Mutation of a phenylalanine residue in the VCP C-terminal tail that can occupy the VAA1 binding site also stimulates ATPase activity, suggesting that VAA1 acts by mimicking this interaction. Together, our findings uncover a druggable allosteric site and a mechanism of enzyme regulation that can be tuned through small molecule mimicry.


Subject(s)
Valosin Containing Protein , Valosin Containing Protein/metabolism , Valosin Containing Protein/chemistry , Valosin Containing Protein/genetics , Allosteric Regulation , Humans , Protein Binding , Molecular Mimicry , Cryoelectron Microscopy , Adenosine Triphosphatases/metabolism , Adenosine Triphosphatases/chemistry , Binding Sites , Allosteric Site , Models, Molecular , Protein Conformation
19.
Proc Natl Acad Sci U S A ; 121(21): e2312755121, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38743628

ABSTRACT

Antigenic similarities between Zika virus (ZIKV) and other flaviviruses pose challenges to the development of virus-specific diagnostic tools and effective vaccines. Starting with a DNA-encoded one-bead-one-compound combinatorial library of 508,032 synthetic, non-natural oligomers, we selected and characterized small molecules that mimic ZIKV epitopes. High-throughput fluorescence-activated cell sorter-based bead screening was used to select molecules that bound IgG from ZIKV-immune but not from dengue-immune sera. Deep sequencing of the DNA from the "Zika-only" beads identified 40 candidate molecular structures. A lead candidate small molecule "CZV1-1" was selected that correctly identifies serum specimens from Zika-experienced patients with good sensitivity and specificity (85.3% and 98.4%, respectively). Binding competition studies of purified anti-CZV1-1 IgG against known ZIKV-specific monoclonal antibodies (mAbs) showed that CZV1-1 mimics a nonlinear, neutralizing conformational epitope in the domain III of the ZIKV envelope. Purified anti-CZV1-1 IgG neutralized infection of ZIKV in cell cultures with potencies comparable to highly specific ZIKV-neutralizing mAbs. This study demonstrates an innovative approach for identification of synthetic non-natural molecular mimics of conformational virus epitopes. Such molecular mimics may have value in the development of accurate diagnostic assays for Zika, as well as for other viruses.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Epitopes , Zika Virus Infection , Zika Virus , Zika Virus/immunology , Epitopes/immunology , Humans , Zika Virus Infection/immunology , Zika Virus Infection/virology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Immunoglobulin G/immunology , Antibodies, Monoclonal/immunology , Molecular Mimicry/immunology
20.
Rev Alerg Mex ; 71(1): 57, 2024 Feb 01.
Article in Spanish | MEDLINE | ID: mdl-38683075

ABSTRACT

OBJECTIVE: Identify molecular mimicry between TPO, eosinophil peroxidase (EPX), thyroglobulin and IL24 and microorganism antigens. METHODS: Through in silico analysis, we performed local alignments between human and microorganism antigens with PSI-BLAST. Proteins that did not present a 3D structure were modeled by homology through the Swiss Modeller server and epitope prediction was performed through Ellipro. Epitopes were located in the 3D models using PYMOL software. RESULTS: A total of 38 microorganism antigens (parasites, bacteria) had identities between 30% and 45%, being the highest with Anisakis simplex. The alignment between 2 candidate proteins from A. simplex and EPX presented significant values, with identities of 43 and 44%. In bacteria, Campylobacter jejuni presented the highest identity with thyroglobulin (35%). 220 linear and conformational epitopes of microorganism antigens were predicted. Peroxidasin-like proteins from Toxocara canis and Trichinella pseudospiralis presented 10 epitopes similar to TPO and EPX, as possible molecules triggering cross-reactivity. No virus presented identity with the human proteins studied. CONCLUSION: TPO and EPX antigens shared potential cross-reactive epitopes with bacterial and nematode proteins, suggesting that molecular mimicry could be a mechanism that explains the relationship between infections and urticaria/hypothyroidism. In vitro work is needed to demonstrate the results obtained in the in silico analysis.


OBJETIVO: Identificar mimetismo molecular entre TPO, eosinofil peroxidasa (EPX), tiroglobulina e IL24 y antígenos de microorganismos. MÉTODOS: A través de análisis in silico, realizamos los alineamientos locales entre los antígenos humanos y de microorganismos con PSI-BLAST. Las proteínas que no presentaban estructura 3D, fueron modeladas por homología a través del servidor Swiss Modeller y se realizó una predicción de epítopes a través de Ellipro. Los epítopes se localizaron en los modelos 3D utilizando el software PYMOL. RESULTADOS: Un total de 38 antígenos de microorganismos (parásitos y bacterias), tuvieron identidades entre 30 y 45%, siendo los más altos con Anisakis simplex. El alineamiento entre dos proteínas candidatas de A. simplex y EPX presentaron valores importantes, con identidades de 43 y 44%. En las bacterias, Campylobacter jejuni presentó la mayor identidad con tiroglobulina (35%). Se predijeron 220 epítopes lineales y conformacionales de antígenos de microorganismos. Las proteínas similares a la peroxidasina de Toxocara canis y Trichinella pseudospiralis presentaron diez epítopes similares a TPO y EPX, como posibles moléculas desencadenantes de una reactividad cruzada. Ningún virus presentó identidad con las proteínas humanas estudiadas. CONCLUSIÓN: Los antígenos TPO y EPX compartieron potenciales epítopes de reacción cruzada con proteínas bacterianas y nematodos, lo que sugiere que el mimetismo molecular podría ser un mecanismo que explique la relación entre infecciones y la urticaria/hipotiroidismo. Se necesitan trabajos in vitro que demuestren los resultados obtenidos en el análisis in silico.


Subject(s)
Autoantigens , Iodide Peroxidase , Molecular Mimicry , Thyroglobulin , Molecular Mimicry/immunology , Humans , Thyroglobulin/immunology , Iodide Peroxidase/immunology , Eosinophil Peroxidase/immunology , Animals , Antigens, Bacterial/immunology , Cross Reactions , Iron-Binding Proteins/immunology , Epitopes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL