Ghofrane Medyouni, Orsolya Vörös, György Panyi, Péter Hajdu
{"title":"Functional role of Kv1.3 localization in chimeric antigen receptor T cells.","authors":"Ghofrane Medyouni, Orsolya Vörös, György Panyi, Péter Hajdu","doi":"10.1093/jimmun/vkaf199","DOIUrl":"https://doi.org/10.1093/jimmun/vkaf199","url":null,"abstract":"<p><p>Chimeric antigen receptor (CAR) T-cell therapy has proven to be a promising treatment for multiple types of cancer. Yet, the mechanisms regulating CAR T-cell function as well as the side effects remain an area of active research. The formation of the immunological synapse is essential for the activation of signaling pathways including the Ca2+-dependent one. Here we demonstrated the functional role of Kv1.3 channels in a CAR T-cell model. Our findings highlight the colocalization of Kv1.3 channel with CAR and its redistribution into the synapse between a CAR T and target cell. The biophysical properties of Kv1.3 channel are not vastly affected by the introduction of CAR in the cells. The blockage of this ion channel's lateral movement affects the killing potential of CAR T cells, likely via disruption of the Ca2+ response upon IS formation. Overall, these data suggest that the manipulation of the Kv1.3 channel may contribute to the improvement of CAR T-cell immunotherapy and provide new insights for future clinical strategies.</p>","PeriodicalId":16045,"journal":{"name":"Journal of immunology","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-08-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144957015","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Xiuping Liang, Yanhong Li, Deying Huang, Xueting Zhao, Yinlan Wu, Ronghui Liao, Tong Wu, Ziyi Tang, Chunyu Tan, Lu Cheng, Yubin Luo, Yi Liu
{"title":"Establishment of a mouse model of Sjögren syndrome-related interstitial lung disease.","authors":"Xiuping Liang, Yanhong Li, Deying Huang, Xueting Zhao, Yinlan Wu, Ronghui Liao, Tong Wu, Ziyi Tang, Chunyu Tan, Lu Cheng, Yubin Luo, Yi Liu","doi":"10.1093/jimmun/vkaf211","DOIUrl":"https://doi.org/10.1093/jimmun/vkaf211","url":null,"abstract":"<p><p>Sjögren syndrome-related interstitial lung disease (SS-ILD) is a severe complication associated with significant morbidity and mortality. Despite its clinical importance, the underlying pathogenesis remains poorly understood, and effective therapeutic strategies are limited. The development of a reliable animal model for SS-ILD is crucial for elucidating disease mechanisms and facilitating the discovery of novel treatments. In this study, we established a SS-ILD mouse model by administering 3 mg/kg bleomycin (BLM) via tracheal exposure to NOD/Ltj mice, a spontaneous model of Sjögren syndrome, with ICR mice serving as controls. The successful induction of Sjögren syndrome was confirmed through histopathology, and SSA/SSB ELISAs. Following BLM administration, lung inflammation and fibrosis were evaluated in ICR and NOD/Ltj mice by imaging, histopathology, and flow cytometry. Both ICR and NOD/Ltj mice developed lung inflammation and fibrosis after BLM exposure. However, NOD/Ltj mice exhibited a more pronounced immune response in the lung, characterized by increased infiltration of immune cells, including monocytes, monocyte-derived macrophages, dendritic cells, neutrophils, T cells, and B cells. This model successfully recapitulates key features of SS-ILD, including concurrent lymphocyte infiltration in the salivary glands and inflammation and fibrosis in the lungs. This study established a novel SS-ILD mouse model replicating human disease pathology, offering a valuable tool for investigating pathogenesis and advancing therapeutic development.</p>","PeriodicalId":16045,"journal":{"name":"Journal of immunology","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-08-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144957024","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Victoria L Palmer, N Max Schabla, Vikas Kumar, Patrick C Swanson
{"title":"RACK1 is required for normal B cell development and signaling but not RAG1 degradation.","authors":"Victoria L Palmer, N Max Schabla, Vikas Kumar, Patrick C Swanson","doi":"10.1093/jimmun/vkaf217","DOIUrl":"https://doi.org/10.1093/jimmun/vkaf217","url":null,"abstract":"<p><p>V(D)J recombination is constrained by timely degradation of the RAG1 and RAG2 proteins through distinct mechanisms. Previously, we showed that full-length RAG1 stability is regulated by viral protein R binding protein (VprBP) through its association with an amino-terminal region in RAG1, but the mechanism remains unclear. As an unbiased approach to uncover potential cofactors involved in the process, we compared protein interactomes between RAG1/RAG2 complexes formed when the amino-terminal third of RAG1 was present or absent. These experiments identified RACK1 as preferentially associating with full-length RAG1. Because RACK1 is implicated in mediating protein degradation in other contexts, we evaluated how loss of RACK1 in B cells affects B cell development and V(D)J recombination. We find that conditional disruption of Rack1 expression in the B lineage in mice blocks B cell development at the pro-B cell stage and impairs V(D)J recombination after Igh DH-JH rearrangement. In this background, enforced Bcl2 expression does not significantly rescue B cell development but does enable the V(D)J recombination defect to be bypassed. However, the phenotype of these mice does not show the excessive Igk rearrangement, skewing toward Igλ+ B cells, or increased RAG1 protein levels observed when VprBP expression is similarly disrupted in B cells, arguing against RACK1 serving as a cofactor in RAG1 degradation. Further studies provide evidence that loss of RACK1 in primary B cells dysregulates cell cycle progression, apoptosis, proliferation, and signaling through MAPK and NF-κB pathways.</p>","PeriodicalId":16045,"journal":{"name":"Journal of immunology","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-08-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12380162/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144957051","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Complement C3 deficiency inhibits osteoclast differentiation and prevents ovariectomy-induced osteoporosis.","authors":"Adrita Guha, Arpita Prasad, Krishna Ashokkumar, Pradipta Pal, Arvind Sahu, Mohan R Wani, Girdhari Lal","doi":"10.1093/jimmun/vkaf180","DOIUrl":"https://doi.org/10.1093/jimmun/vkaf180","url":null,"abstract":"<p><p>The pathomechanistic role of the complement system is well recognized in various pathological conditions affecting bone tissues and the bone microenvironment, including rheumatoid arthritis, osteoarthritis, bone fractures, and periodontitis. The homeostasis of the bone is maintained by continuous remodeling, in which bone-resorbing or demineralizing osteoclast cells remove bone calcification, and osteoblast cells deposit new bone matrix. Major complement protein C3 is reported to control endochondral ossification, cartilage-to-bone transition, and longitudinal bone growth. The role of the complement protein C3 in differentiating multinucleated osteoclast cells (bone-resorbing cells) from osteoclast precursor cells (OCPs) and its contribution to long bone microarchitecture and strength are unclear. We demonstrated that C3 promotes the differentiation of osteoclasts and the formation of multinucleated osteoclasts from bone marrow-derived OCPs. C3-/- mice OCPs had reduced osteoclast-associated gene expression of TRAP (tartrate-resistant acid phosphatase), cathepsin K, calcitonin receptor, and RANK (receptor activator of nuclear factor κB) molecules compared with osteoclasts derived from wild-type (WT) OCPs. C3-/- mice had significantly increased bone mineral density and other bone parameters of the femur compared with WT mice. Furthermore, compared with WT mice, C3-/- mice were protected from ovariectomy-induced osteoporosis, characterized by significantly increased Foxp3+CD4+ T cells in the spleen and interleukin-10-producing B cells in both the spleen and the bone marrow. Intriguingly, C3 knockout mice exhibit reduced differentiation of functional osteoclast cells, which promotes a strong bone microarchitecture, suggesting that complement pathways may be explored as a therapeutic target in bone inflammatory diseases.</p>","PeriodicalId":16045,"journal":{"name":"Journal of immunology","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-08-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144957048","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Magali M Moretto, Keer Chen, Christina Cox, Jie Chen, Imtiaz A Khan
{"title":"In a model of parasite-mediated exhaustion, stem-like CD8 T cells differentiate into an unconventional intermediate effector memory subset.","authors":"Magali M Moretto, Keer Chen, Christina Cox, Jie Chen, Imtiaz A Khan","doi":"10.1093/jimmun/vkaf165","DOIUrl":"https://doi.org/10.1093/jimmun/vkaf165","url":null,"abstract":"<p><p>CD8 T cell exhaustion has been reported in mice susceptible to Toxoplasma gondii infection. While the differentiation of CD8 exhausted subsets has been extensively reported, most of these studies have been conducted in chronic viral and cancer models. During chronic T. gondii infection, phenotypic and transcriptomic analyses of the polyclonal antigen-specific CD8 T cell response characterize 4 populations based on KLRG1 and CD62L expression. Pop1 (KLRG1+CD62Llo) bears the attributes of a terminal effector subset, and pop2 (KLRG1-CD62Llo) is similar to effector memory CD8 T cells. Akin to chronic viral infection and cancer systems, pop3 (KLRG1-CD62Lhi) exhibits the characteristics of stem-like progenitor CD8 T cells (high Tcf7, Slamf6, and Cxcr5 expression), whereas pop4 (KLRG1+CD62Lhi) closely resembles a transitory subset (elevated Tbx21, low Tcf1, and Tox expression). During chronic viral infection, the stem-like progenitor CD8 T cells transition into a terminally differentiated exhausted subset via an intermediate population. However, in our system, pop3 (KLRG1-CD62Lhi) generates pop4 (KLRG1+CD62Lhi), which does not convert into a conventional terminally differentiated exhausted subset but instead transitions into effector pop1 (KLRG1+CD62Llo). Notably, during the chronic phase of the infection, pop1 cannot retain its functionality, irrespective of its origin, which may hamper its ability to control reactivation. Our observations emphasize that the differentiation of exhausted CD8 T cells in non-viral infections, like chronic toxoplasmosis, follows a different pattern than established models and highlights the need to develop new immune strategies better tailored for a broad range of pathogens.</p>","PeriodicalId":16045,"journal":{"name":"Journal of immunology","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-08-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144957062","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Connor W McKaig, Jill Malfetano, Y Tran, Xiuli Yang, Utpal Pal, Keith Wycoff, Yi-Pin Lin
{"title":"Complement therapeutic factor H-IgG proteins as pre-exposure prophylaxes against Lyme borreliae infections.","authors":"Connor W McKaig, Jill Malfetano, Y Tran, Xiuli Yang, Utpal Pal, Keith Wycoff, Yi-Pin Lin","doi":"10.1093/jimmun/vkaf195","DOIUrl":"10.1093/jimmun/vkaf195","url":null,"abstract":"<p><p>Lyme disease (LD) is the most common vector-borne disease in the northern hemisphere and is caused by the bacteria Borrelia burgdorferi sensu lato (also known as Lyme borreliae) with no effective prevention available. Lyme borreliae evade complement killing, a critical arm of host immune defense, by producing outer surface proteins that bind to a host complement inhibitor, factor H (FH). These outer surface proteins include CspA and CspZ, which bind to the sixth and seventh short consensus repeats (SCR6-7) of FH and the OspE family of proteins (OspE), which bind to the nineteenth and twentieth SCR (SCR19-20). In this study, we produced 2 chimeric proteins, FH-Fc, containing the Fc region of immunoglobulin G (Fc) with SCR6-7 or SCR19-20. We found that both FH-Fc constructs killed B. burgdorferi via bacterial lysis and phagocytosis and reduced bacterial colonization and LD-associated joint inflammation in vivo. While SCR6-7-Fc displayed Lyme borreliae species-specific bacterial killing, SCR19-20-Fc versatilely eradicated all tested bacterial species/strains. This correlated with SCR6-7-Fc binding to select variants of CspA and CspZ, but SCR19-20-Fc binding to all tested OspE variants. Overall, we demonstrated the concept of using FH-Fc constructs to kill Lyme borreliae and defined underlying mechanisms, highlighting the potential of FH-Fc as a pre-exposure prophylaxis against LD infection.</p>","PeriodicalId":16045,"journal":{"name":"Journal of immunology","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-08-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12435995/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144957053","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Janyerkye Tulyeu, David Priest, James B Wing, Jonas Nørskov Søndergaard
{"title":"Optimized detection and inference of immune cell type names in single-cell RNA sequencing data.","authors":"Janyerkye Tulyeu, David Priest, James B Wing, Jonas Nørskov Søndergaard","doi":"10.1093/jimmun/vkaf183","DOIUrl":"https://doi.org/10.1093/jimmun/vkaf183","url":null,"abstract":"<p><p>Accurate identification of immune cell subsets in single-cell RNA sequencing (scRNA-seq) data is critical for understanding immune responses in autoimmune diseases, infections, and cancer. One caveat of scRNA-seq is the inability to properly assign rare immune cell subsets due to gene dropout events. To circumvent this caveat, we here developed optimized detection and inference of names in scRNA-seq data (scODIN). scODIN uses an informed holistic 2-step approach combining expert knowledge with machine learning to rapidly assign cell type identities to large scRNA-seq datasets. First, scODIN uses key lineage-defining markers to identify a set of core cell types. Second, scODIN compensates for dropout events by integrating a k-nearest neighbors algorithm. We additionally programmed scODIN to detect dual and transitional phenotypes, which are usually overlooked in conventional analyses. Consequently, scODIN may enhance our understanding of immune cell heterogeneity and provide comprehensive insights into immune regulation, with broad implications for immunology and personalized medicine.</p>","PeriodicalId":16045,"journal":{"name":"Journal of immunology","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-08-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144957068","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Philip Barbulescu, Matthew K Wong, Leon Baronian, Pengyu Wang, Abdulmateen Aderinto, Matthias Kneussel, Alberto Martin
{"title":"MKLN1-dependent GID4/CTLH E3 ubiquitin ligase complex assemblies are required to support B-cell antibody diversification.","authors":"Philip Barbulescu, Matthew K Wong, Leon Baronian, Pengyu Wang, Abdulmateen Aderinto, Matthias Kneussel, Alberto Martin","doi":"10.1093/jimmun/vkaf201","DOIUrl":"https://doi.org/10.1093/jimmun/vkaf201","url":null,"abstract":"<p><p>C-terminal to LisH (CTLH) E3 ubiquitin ligase complexes regulate a broad range of biological processes and forms separate supramolecular CTLH-MKLN1 and CTLH-WDR26 assemblies possessing distinct substrate specificities. Our previous work revealed that the CTLH complex utilizes the FAM72A substrate adaptor to ubiquitinate and degrade the uracil-DNA glycosylase 2 (UNG2) base excision repair factor. This outcome in B cells permits deoxyuridine mutations catalyzed by activation-induced cytidine deaminase (AID) to persist toward mutational outcomes and drive antibody diversification events. Here, we report that Mkln1-/- mice specifically lacking assembly of CTLH-MKLN1 complexes display reduced somatic hypermutation and class switch recombination frequencies due to increased UNG2, similar to Fam72a-/- mice. Strikingly, Mkln1-/- mice showed increased germinal center B cells and defects during B-cell development, a phenotype not observed in Fam72a-/- mice, suggesting that MKLN1 regulates proteins that are independent of FAM72A. Together, this work identifies that CTLH-MKLN1 ubiquitin E3 ligase complexes are critical in generating effective humoral immune responses and reveals distinctions between FAM72A-dependent and -independent CTLH complex modalities.</p>","PeriodicalId":16045,"journal":{"name":"Journal of immunology","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-08-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144956994","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Akhilesh Kumar, Bin Zhang, Andrew Baldys, Colin Fischer, Alexander Lenvik, Martin Felices, Zachary B Davis, Laura Bendzick, Anna J Weis, Yenan T Bryceson, Jeffrey S Miller, Frank Cichocki
{"title":"The transcription factor BCL11B drives NK cell cytotoxicity and antitumor activity.","authors":"Akhilesh Kumar, Bin Zhang, Andrew Baldys, Colin Fischer, Alexander Lenvik, Martin Felices, Zachary B Davis, Laura Bendzick, Anna J Weis, Yenan T Bryceson, Jeffrey S Miller, Frank Cichocki","doi":"10.1093/jimmun/vkaf179","DOIUrl":"https://doi.org/10.1093/jimmun/vkaf179","url":null,"abstract":"<p><p>Bcl11b is a zinc-finger transcription factor that is required for the differentiation of αβ T cells. A role for BCL11B in the regulation of human natural killer (NK) cell maturation has been inferred from patients with heterozygous mutations in BCL11B. However, mechanistic and functional studies are lacking due to the low efficiency of current transduction and transfection methods. Here, we developed a synthetic BCL11B mRNA with enhanced stability that could be transfected into primary NK cells at high frequencies. Introduction of BCL11B mRNA slowed NK cell proliferation while simultaneously driving maturation and acquisition of cytotoxic granule components. For in vitro and in vivo functional testing, we generated induced pluripotent stem cell (iPSC)-derived NK (iNK) cells with inducible BCL11B expression. These iNK cells mediated faster solid tumor cell killing and significantly better tumor control in vivo. Together, our findings support the notion that BCL11B is a key transcription factor in human NK cells and demonstrate that increased BCL11B expression can enhance NK cell immunotherapy.</p>","PeriodicalId":16045,"journal":{"name":"Journal of immunology","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-08-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144956989","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ningxia Xiong, Maolin Lv, Jingjing Zhang, Yuezong Xu, Bo Tang, Chunrong Yang, Jianguo Su
{"title":"TLR4al senses heme as a key damage/danger-associated molecular pattern to activate immune responses in lower vertebrates.","authors":"Ningxia Xiong, Maolin Lv, Jingjing Zhang, Yuezong Xu, Bo Tang, Chunrong Yang, Jianguo Su","doi":"10.1093/jimmun/vkaf192","DOIUrl":"10.1093/jimmun/vkaf192","url":null,"abstract":"<p><p>Toll-like receptor 4 (TLR4), a critical pattern recognition receptor, detects microbe- and damage/danger-associated molecular patterns to trigger immune responses in mammals. However, the functions and mechanisms remain largely unclear in lower vertebrates. This study systematically investigates the evolutionary divergence, subcellular localization and ligand of TLR4 in lower vertebrates by grass carp (Ctenopharyngodon idella) as a model species. TLR4 emerges and expands in a few teleosts but is absent in most fishes. We standardized the nomenclature of multiple TLR4 variants (3 or 4) in cyprinids. Tetrapods generally contain 1 TLR4. Astonishingly, CiTLR4ba, CiTLR4al, and CiTLR4bb localize to lysosomes but not cytomembrane like their mammalian counterparts, in which they recognize ligands that are engulfed into lysosomes. However, CiTLR4bc is soluble in cytoplasm due to the absence of signal peptide and transmembrane domain. In ligand recognition, CiTLR4 variants exhibit obvious heterogeneity. CiTLR4bc exhibits no binding activity to any of the tested ligands [lipopolysaccharide, peptidoglycan, bacterial dsDNA, polyinosinic-polycytidylic acid, heme]. Interestingly, both CiTLR4ba and CiTLR4bb bind polyinosinic-polycytidylic acid. Excitedly, CiTLR4al conservatively recognizes heme, a central damage/danger-associated molecular pattern (DAMP) released from damaged erythrocytes, in teleost and human. Further, 5 key high-affinity heme-binding sites (C113, H185, C295, H342, and C458) were identified in CiTLR4al. Each single key site mutation attenuates the heme-binding ability and downstream immune responses of CiTLR4al, supporting a multivalent heme-CiTLR4al interaction mechanism. The present study systematically elucidates the evolution of TLR4, as well as its complexity and conservation in vertebrates, offering direct evidence for the adaptive evolution of TLR4 in vertebrates. The results also contribute to the immune mechanism of hemolytic diseases.</p>","PeriodicalId":16045,"journal":{"name":"Journal of immunology","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-08-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144883009","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}