Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,095)

Search Parameters:
Keywords = T cell persistence

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
9 pages, 221 KB  
Case Report
Therapy-Related Myeloid Neoplasms After CAR-T Therapy: A Case Series with Distinct Cytogenetic Features and Comparison with Autologous Stem Cell Transplantation
by Pilar Palomo-Moraleda, Sara Alonso-Álvarez, Lucía Morais-Bras, Christian Sordo-Bahamonde, Rocío Granda-Díaz, Joud Zanabili-Al-Sibai, Sofía García-Ferreiro, Marco Moro-García, Estefanía Pérez-López, Marco Hernández-Martín, Ana J. González-Huerta, Soledad González-Muñiz, Ángel Ramírez-Payer, J. María García-Gala, Ariana Fonseca-Mourelle, Segundo González and Ana P. González-Rodríguez
Hemato 2026, 7(1), 1; https://doi.org/10.3390/hemato7010001 - 25 Dec 2025
Abstract
Background: The emergence of therapy-related myelodysplastic syndrome (t-MN) after autologous stem cell transplantation (ASCT) is well documented. However, with the growing use of chimeric antigen receptor (CAR) T-cell therapy for relapsed/refractory B-cell malignancies, concerns about secondary myeloid neoplasms, particularly MN, have arisen. The [...] Read more.
Background: The emergence of therapy-related myelodysplastic syndrome (t-MN) after autologous stem cell transplantation (ASCT) is well documented. However, with the growing use of chimeric antigen receptor (CAR) T-cell therapy for relapsed/refractory B-cell malignancies, concerns about secondary myeloid neoplasms, particularly MN, have arisen. The mechanisms and cytogenetic features associated with post-CAR-T MN, especially chromosome 7 abnormalities, remain underexplored. Objectives: To compare the incidence, timing, and cytogenetic characteristics of MN developing after CAR-T-cell therapy versus ASCT, and to evaluate the potential association between CAR-T therapy, persistent cytopenias, and these specific alterations. Study Design: This was a retrospective, single-center study of 275 patients with B-cell malignancies treated between 2015 and 2024 at Hospital Universitario Central de Asturias (Spain). Of these, 259 patients underwent ASCT and 16 received CAR-T-cell therapy (axicabtageneciloleucel n = 13, tisagenlecleucel n = 2, brexucabtageneautoleucel n = 1). Clinical, cytogenetic, and laboratory data were collected and analyzed. Incidence rates were compared using Fisher’s exact test, and time-to-event outcomes was evaluated using the Mann–Whitney U test (given the small number of events). Statistical significance was set at p < 0.05. Results: Myeloid neoplasms were diagnosed in 3 of 259 ASCT patients (1.15%) and in 2 of 16 CAR-T-cell patients (12.5%) (p = 0.03). The median time to myeloid neoplasm diagnosis was numerically shorter in the CAR-T group (15.5 vs. 69 months, p = 0.096). All post-CAR-T cases presented persistent cytopenias and cytokine release syndrome (CRS). Cytogenetic analyses revealed de novo monosomy 7 and 7q deletion in both CAR-T-related cases, whereas no chromosome 7 abnormalities were detected in ASCT-related cases. Pre-treatment samples did not show these abnormalities, although limitations in the sensitivity of the assays preclude the definitive exclusion of minor pre-existing clones. Both affected CAR-T patients had prolonged CAR-T cell persistence and required transfusional support due to hematologic toxicity. One patient was diagnosed with high-risk MN with 5q and 7q deletion and the other with Clonal Cytopenia of Uncertain Significance (CCUS) with monosomy 7. Conclusions: CAR-T-cell therapy was associated with a significantly higher and earlier incidence of myeloid neoplasms compared to ASCT in this cohort. The development of post-CAR-T myeloid neoplasm was characterized by persistent cytopenias, prolonged CAR-T cell persistence, and de novo chromosome 7 alterations. While the small sample size necessitates cautious interpretation, these findings may suggest a distinct pathogenesis potentially linked to inflammation, immune toxicity, or the expansion of pre-existing clones. This highlights the need for long-term hematologic monitoring and evaluation for clonal hematopoiesis prior to CAR-T-cell therapy, especially in heavily pretreated patients. Full article
22 pages, 1501 KB  
Review
The Convergent Immunopathogenesis of Cigarette Smoke Exposure: From Oxidative Stress to Epigenetic Reprogramming in Chronic Disease
by Aysen Kutan Fenercioglu, Hafize Uzun and Durisehvar Ozer Unal
Int. J. Mol. Sci. 2026, 27(1), 187; https://doi.org/10.3390/ijms27010187 - 24 Dec 2025
Viewed by 57
Abstract
Cigarette smoking is the leading preventable cause of chronic diseases (e.g., COPD, cardiovascular disease, cancer), largely driven by persistent immune-inflammatory mechanisms. This review synthesizes the molecular and cellular cascades linking cigarette smoke (CS) exposure to chronic pathology. CS constituents, particularly ROS/RNS, induce rapid [...] Read more.
Cigarette smoking is the leading preventable cause of chronic diseases (e.g., COPD, cardiovascular disease, cancer), largely driven by persistent immune-inflammatory mechanisms. This review synthesizes the molecular and cellular cascades linking cigarette smoke (CS) exposure to chronic pathology. CS constituents, particularly ROS/RNS, induce rapid oxidative stress that overwhelms antioxidant defenses and generates damage-associated molecular patterns (DAMPs). These DAMPs activate pattern recognition receptors (PRRs) and the NLRP3 inflammasome, initiating NF-κB signaling and the release of pro-inflammatory cytokines (TNF-α, IL-1β, IL-6). CS exposure causes profound innate immune dysregulation, including airway epithelial barrier disruption, hyperactivated neutrophils, and dysfunctional alveolar macrophages (AMs) that release destructive proteases (e.g., MMP-12) and acquire foam-cell–like characteristics. Furthermore, CS drives adaptive immunity toward a Th1/Th17-dominant phenotype while suppressing regulatory T-cell (Treg) function, thereby promoting autoimmunity and chronic tissue injury. Critically, CS induces epigenetic reprogramming (e.g., DNA methylation, miRNA dysregulation), locking immune cells into a persistent pro-inflammatory state. This convergence of oxidative stress, innate and adaptive immune dysregulation, and epigenetic alterations underlies the systemic low-grade inflammation that fuels smoking-related chronic diseases, highlighting key targets for novel therapeutic interventions. Full article
Show Figures

Graphical abstract

18 pages, 6505 KB  
Article
Pre-Treatment with Dacarbazine Sensitizes B16 Melanoma to CAR T Cell Therapy in Syngeneic Mouse Model
by Egor A. Emelianov, Elizaveta R. Naberezhnaya, Andrey S. Logvinov, Valeria M. Stepanova, Aleksandr S. Chernov, Yuliana A. Mokrushina, Diana M. Malabuiok, Dmitry E. Pershin, Ekaterina A. Malakhova, Elena A. Kulakovskaya, Tatiana N. Prokofeva, Victor V. Tatarskiy, Elena I. Shramova, Sergey M. Deyev, Alexander G. Gabibov, Nikolay E. Kushlinskii, Yury P. Rubtsov and Dmitry V. Volkov
Int. J. Mol. Sci. 2026, 27(1), 189; https://doi.org/10.3390/ijms27010189 - 24 Dec 2025
Viewed by 82
Abstract
Adoptive cell therapy (ACT) with T cells modified with a chimeric antigen receptor (CAR T cells) has dramatically improved outcomes in hematologic cancers. However, its efficacy in solid tumors, such as melanoma, is hampered by several factors. These include heterogeneous expression of tumor-associated [...] Read more.
Adoptive cell therapy (ACT) with T cells modified with a chimeric antigen receptor (CAR T cells) has dramatically improved outcomes in hematologic cancers. However, its efficacy in solid tumors, such as melanoma, is hampered by several factors. These include heterogeneous expression of tumor-associated antigens (TAA) and an immunosuppressive, profibrotic tumor microenvironment (TME), which restricts cytotoxic CAR T cells trafficking into the tumor, as well as their persistence and cytolytic activity. As a result, responses to CAR T cell monotherapy in melanoma and other solid tumors are typically weak, transient or even absent. Emerging evidence suggests that combining traditional chemotherapy with CAR T cell therapy can enhance the antitumor activity of CAR T cells in solid malignancies. Partial tumor cell killing by chemotherapy improves access to TAA and disrupts the TME by affecting the global structure of the tumor tissue. Here, we developed an immunocompetent syngeneic B16 melanoma mouse model to test a combination of classical dacarbazine (DTIC) chemotherapy with ACT with murine CAR T cells. B16-F10 (next as B16) melanoma cells were modified to express a human/murine hybrid epidermal growth factor receptor (EGFR) recognized by a murine CAR bearing a single-chain variable fragment (scFv) derived from cetuximab, an anti-EGFR monoclonal antibody approved for the treatment of colorectal and certain other solid tumors. Prior to CAR T cells administration, cyclophosphamide (CPA) pre-conditioning was used. We demonstrated that DTIC therapy followed by infusion of murine CAR T cells targeting the human/murine hybrid EGFR (EGFR mCAR T cells) provided superior tumor control and prolonged survival compared to monotherapy with either DTIC or EGFR mCAR T cells alone. These findings support the potential feasibility of a combined therapeutic strategy for human melanoma involving DTIC treatment followed by EGFR CAR T cells infusion after CPA pre-conditioning. Full article
(This article belongs to the Special Issue Chimeric Antigen Receptors Against Cancers and Autoimmune Diseases)
Show Figures

Graphical abstract

14 pages, 1779 KB  
Article
Pilot Proteomic Analysis of Urinary Extracellular Vesicles Supports the “Toxic Urine Hypothesis” as a Vicious Cycle in Refractory IC/BPS Pathogenesis
by Man-Jung Hung, Evelyn Yang, Tsung-Ho Ying, Peng-Ju Chien, Ying-Ting Huang and Wen-Wei Chang
Int. J. Mol. Sci. 2026, 27(1), 130; https://doi.org/10.3390/ijms27010130 - 22 Dec 2025
Viewed by 329
Abstract
Despite treatments such as pentosan polysulfate, hyaluronic acid, botulinum toxin A, and platelet-rich plasma, many interstitial cystitis/bladder pain syndrome (IC/BPS) patients experience persistent symptoms. Urinary extracellular vesicles (uEVs) carry molecular cargo reflecting disease pathophysiology, yet their proteomic profiles in treated IC/BPS remain unexplored. [...] Read more.
Despite treatments such as pentosan polysulfate, hyaluronic acid, botulinum toxin A, and platelet-rich plasma, many interstitial cystitis/bladder pain syndrome (IC/BPS) patients experience persistent symptoms. Urinary extracellular vesicles (uEVs) carry molecular cargo reflecting disease pathophysiology, yet their proteomic profiles in treated IC/BPS remain unexplored. This pilot study examined uEV proteomics in refractory IC/BPS cases to test the “Toxic Urine Hypothesis”—a vicious cycle, whereby urothelial dysfunction enables EV-mediated toxin penetration, triggering inflammation that further impairs the bladder barrier. Urinary EVs were isolated from six female IC/BPS patients on active treatments and four healthy female controls. Mass spectrometry-based proteomics identified differential protein expressions, followed by pathway enrichment analysis and functional validation using NF-κB reporter assays in HEK293T cells and Western blot in primary human bladder epithelial cells. IC/BPS EVs exhibited 31 upregulated proteins (including HPGD, KRT8, HSPA4, 14-3-3 family members) and 19 downregulated proteins (including neutrophil granule proteins MPO and ELANE), indicating suppressed acute neutrophil inflammation but enriched homeostatic, metabolic, and regenerative pathways. Patient EVs induced significantly higher NF-κB activation than in the controls, with upregulated 14-3-3ζ and phosphorylated NF-κB p65 in bladder epithelial cells. These findings support the “Toxic Urine Hypothesis”, revealing persistent NF-κB-mediated chronic epithelial stress despite suppressed acute inflammation in treated IC/BPS patients, suggesting that therapies targeting inflammation and regeneration may help break this vicious cycle. Full article
(This article belongs to the Special Issue Role of Extracellular Vesicles in Diseases)
Show Figures

Graphical abstract

20 pages, 360 KB  
Review
Alterations in the Immune Response in Individuals with Latent Tuberculosis Infection
by Anna Starshinova, Adilya Sabirova, Igor Kudryavtsev, Artem Rubinstein, Arthur Aquino, Leonid P. Churilov, Ekaterina Belyaeva, Anastasia Kulpina, Raul A. Sharipov, Ravil K. Tukfatullin, Nikolay Nikolenko and Dmitry Kudlay
Pathogens 2026, 15(1), 14; https://doi.org/10.3390/pathogens15010014 - 22 Dec 2025
Viewed by 127
Abstract
Latent tuberculosis infection (LTBI) represents a biologically active yet clinically asymptomatic stage of Mycobacterium tuberculosis (Mtb) persistence. This condition is characterized by subtle immunometabolic alterations reflecting the host–pathogen equilibrium. Understanding the mechanisms and biomarkers associated with the preclinical phase of LTBI is crucial [...] Read more.
Latent tuberculosis infection (LTBI) represents a biologically active yet clinically asymptomatic stage of Mycobacterium tuberculosis (Mtb) persistence. This condition is characterized by subtle immunometabolic alterations reflecting the host–pathogen equilibrium. Understanding the mechanisms and biomarkers associated with the preclinical phase of LTBI is crucial for preventing progression to active tuberculosis (ATB). Recent advances have identified multiple immunological, transcriptomic, metabolic, and imaging-based approaches that enable stratification of individuals at increased risk of LTBI reactivation. Quantitative assays such as IGRA, multiplex and T-cell activation marker (TAM) tests, as well as interferon-related transcriptional signatures, demonstrate predictive potential when combined with functional assays (MGIA) and metabolic imaging (PET/CT). Experimental primate models faithfully reproduce the spectrum from latency to reactivation, allowing for validation of biomarkers and vaccine or immunomodulatory strategies. The review also highlights the particular challenges of multidrug-resistant LTBI (MDR-LTBI), where standard chemoprophylaxis is less effective and immune control plays a decisive role. The preclinical phase of LTBI constitutes a key point in the TB control cascade. Integrating immunological, transcriptomic, and radiological data into risk-based screening algorithms could substantially improve early detection and targeted prevention. Translating research-derived signatures into clinically applicable, standardized, and cost-effective diagnostic tools requires coordinated international efforts, technological transfer, and policy-level support to reduce TB reactivation and transmission, including MDR-TB. Full article
(This article belongs to the Special Issue Innate Immune Response and Pathogen Dynamics)
28 pages, 1074 KB  
Review
CAR-NK Engineering to Overcome TME Barriers
by Fahmida Islam, Aleta Pupovac, Richard L. Boyd and Alan O. Trounson
Cells 2026, 15(1), 21; https://doi.org/10.3390/cells15010021 - 22 Dec 2025
Viewed by 298
Abstract
Chimeric antigen receptor (CAR)-based immunotherapy has shown considerable promise in cancer treatment by redirecting immune effector cells to recognize and eliminate tumor cells in an antigen-specific manner. While CAR-T cells bearing tumor-specific CARs have shown remarkable success in treating some hematological malignancies, their [...] Read more.
Chimeric antigen receptor (CAR)-based immunotherapy has shown considerable promise in cancer treatment by redirecting immune effector cells to recognize and eliminate tumor cells in an antigen-specific manner. While CAR-T cells bearing tumor-specific CARs have shown remarkable success in treating some hematological malignancies, their clinical application is limited by cytokine release syndrome, neurotoxicity, and graft-versus-host disease. In contrast, CAR–natural killer (NK) cells retain their multiple forms of natural anti-tumor capabilities without the pathological side effects and are compatible with allogeneic “off-the-shelf” application by not requiring prior activation signaling. Despite CAR-NK therapies showing promising results in hematological malignancies, they remain limited as effector cells against solid tumors. This is primarily due to the complex, immunosuppressive tumor microenvironment (TME), characterized by hypoxia, nutrient depletion, lactate-induced acidosis, and inhibitory soluble factors. Collectively, these significantly impair NK cell functionality. This review examines challenges faced by CAR-NK therapy in combating solid tumors and outlines strategies to reduce them. Barriers include tumor antigen heterogeneity, immune escape, trogocytosis-mediated fratricide, rigid structural and metabolic barriers in the TME, immunosuppressive factors, and defective homing and cell persistence of CAR-NK cells. We also emphasize the impact of combining other complementary immunotherapies (e.g., multi-specific immune engagers and immunomodulatory agents) that further strengthen CAR-NK efficacy. Finally, we highlight critical research gaps in CAR-NK therapy and propose that cutting-edge technologies are required for successful clinical translation in solid tumor treatment. Full article
(This article belongs to the Special Issue Novel Insights into Cancer Immune Responsiveness)
Show Figures

Graphical abstract

18 pages, 1445 KB  
Review
ADA1-Driven Metabolic Refueling Enhances CAR T Cell Therapy for Solid Tumors
by Alex Wade Song and Xiaotong Song
Cancers 2026, 18(1), 34; https://doi.org/10.3390/cancers18010034 - 22 Dec 2025
Viewed by 152
Abstract
CAR T cell therapy, while highly effective for hematological malignancies, continues to face significant hurdles in the treatment of solid tumors. Key challenges include severe nutrient deprivation and the presence of immunosuppressive metabolites such as adenosine in the tumor microenvironment, which limit CAR [...] Read more.
CAR T cell therapy, while highly effective for hematological malignancies, continues to face significant hurdles in the treatment of solid tumors. Key challenges include severe nutrient deprivation and the presence of immunosuppressive metabolites such as adenosine in the tumor microenvironment, which limit CAR T cell persistence and antitumor activity. This review focuses on current progress and future directions for ADA1-based metabolic reprogramming as a targeted approach to enhance CAR T cell function. We discuss recent advances, particularly the engineering of CAR T cells to express ADA1, which facilitates the local conversion of immunosuppressive adenosine into inosine, thereby supporting T cell metabolism and improving therapeutic outcomes. Preclinical studies, including our own, demonstrate that ADA1-expressing CAR T cells exhibit reduced exhaustion, greater metabolic flexibility, and enhanced antitumor efficacy in solid tumor models. The selective clearance of adenosine and supplementation of inosine directly address the metabolic barriers within the tumor microenvironment and provide an effective strategy to bolster CAR T cell responses. Integration of ADA1-driven metabolic refueling with future innovations in CAR design holds promise for overcoming key obstacles in solid tumor immunotherapy. We conclude by highlighting the potential of ADA1-based strategies and offering our perspective on their translation toward clinical application. Full article
Show Figures

Figure 1

11 pages, 1225 KB  
Article
Development of Approaches for Transgene Expression in the Pathogenic Free-Living Amoeba Naegleria fowleri
by Caroline M. Palmentiero, Jillian E. M. McKeon, Colm P. Roster and James C. Morris
Pathogens 2026, 15(1), 12; https://doi.org/10.3390/pathogens15010012 - 22 Dec 2025
Viewed by 124
Abstract
The absence of molecular tools for manipulation of gene expression in the pathogenic free-living amoeba Naegleria fowleri has historically limited our understanding of gene function in the organism and has coincidently impacted the identification of potential druggable pathways and proteins. Here, we describe [...] Read more.
The absence of molecular tools for manipulation of gene expression in the pathogenic free-living amoeba Naegleria fowleri has historically limited our understanding of gene function in the organism and has coincidently impacted the identification of potential druggable pathways and proteins. Here, we describe the development of approaches for the generation of transgenic amoebae using polyethyleneimine nanoparticles to deliver plasmids designed to confer antibiotic resistance and fluorescence to the cells. Through a series of optimization steps, we found that transfection of plasmids encoding the fluorescent protein mCherry fused by a T2A self-cleaving peptide to a codon-optimized puromycin acetyltransferase selectable marker yielded fluorescent cells that were resistant up to 100 µg/mL puromycin. Transfected trophozoites harbored between 45 and 65 copies of the transgene per cell and both fluorescence and resistance were persistent in the presence of selector through continued passages. The development of these approaches is anticipated to enable application of an array of genetic manipulation techniques including forward and reverse genetics to the study of this important pathogen. Full article
Show Figures

Figure 1

13 pages, 3450 KB  
Article
ROMO1 as a Diagnostic Biomarker in Cervical Neoplasia: Evidence from Normal, Pre-Invasive, and Invasive Lesions
by Eva Tsoneva, Polina Damyanova, Metodi V. Metodiev, Velizar Shivarov, Mariela Vasileva-Slaveva, Zornitsa Gorcheva, Yonka Ivanova, Yavor Kornovski, Stoyan Kostov, Stanislav Slavchev, Margarita Nikolova, Angel Yordanov and Rafał Watrowski
Diagnostics 2026, 16(1), 24; https://doi.org/10.3390/diagnostics16010024 - 21 Dec 2025
Viewed by 154
Abstract
Background: Cervical cancer (CC) is the fourth most common malignancy in women around the world, with more than 600,000 new cases registered in 2022 and around 350,000 deaths. It is a growing social problem, especially in developing countries. Almost all cases of [...] Read more.
Background: Cervical cancer (CC) is the fourth most common malignancy in women around the world, with more than 600,000 new cases registered in 2022 and around 350,000 deaths. It is a growing social problem, especially in developing countries. Almost all cases of cervical cancer are caused by persistent infection with oncogenic high-risk human papillomavirus (HPV). This malignancy usually exhibits a gradual development through well-defined precursor stages, known as cervical intraepithelial neoplasia (CIN) grades 1, 2, and 3, before evolving into invasive carcinoma. In diagnostic practice, several biomarkers have been implemented to improve the detection of high-risk cervical lesions. p16 and Ki-67 greatly aid in identifying HPV-driven dysplasia, but they cannot always reliably distinguish progressive lesions from regressive or transient HPV infections. These limitations highlight the need for novel biomarkers with better predictive accuracy to complement current screening and diagnostic algorithms. ROMO1 has become a possible marker of a high-ROS, high-risk tumor phenotype in a number of cancers. Although oxidative stress, HPV, and cervical carcinogenesis have been linked, nothing is known about ROMO1’s involvement in cervical neoplasia. There is currently a lack of thorough information regarding the expression of ROMO1 in normal vs. precancerous lesions and in cervical cancer, as well as on whether or not its expression is correlated with the severity of the disease. In order to define ROMO1 expression throughout the course of cervical squamous neoplastic development, the current study was created. Methods: We performed immunohistochemical analysis of ROMO1 expression on cervical tissue samples from three groups: healthy cervix (n = 30), cervical intraepithelial neoplasia (CIN) (n = 41), and invasive cervical carcinoma (n = 205). ROMO1 expression in invasive carcinoma was evaluated using an H-score scale. Results: ROMO1 expression was basal in all normal cervix samples (0/30 cases). In contrast, CIN lesions showed 100% ROMO1 expression in the suprabasal layers of abnormal cells in all CIN cases. In invasive cervical carcinomas, ROMO1 expression was heterogeneous. In our cancer cohort (n = 205), ROMO1 H-score showed no significant association with the following: FIGO stage I vs. II vs. III (p = 0.25); histologic grade G1 vs. G2 vs. G3 (p = 0.46); lymphovascular invasion (no vs. yes; p = 0.80); nodal status N0 vs. N1 (p = 0.67); patient age (≤50 y vs. >50 y; p = 0.38). However, ROMO1 expression did vary by histologic subtype (AC vs. ASC vs. SCC; p = 0.02), with SCC enriched for strong staining compared to AC/ASC. With regard to tumor stage (pT stage), pT2a tumors exhibited significantly lower ROMO1 (pT1b1–pT2b; p = 0.035) than pT1b1 (p = 0.04). No other clinicopathologic variable remained significant. Notably, ROMO1 expression was highest in stage I tumors and declined in more advanced stages of cervical carcinoma. Conclusions: These results show a clear pattern of ROMO1 expression across the cervical neoplasia spectrum: it is attenuated in invasive tumors (with a peak in early-stage illness), significantly raised in pre-cancerous CIN lesions, and negligible in normal epithelium. The idea that oxidative stress may be the primary cause of early malignant transformation in the cervix is supported by the noticeable overexpression of ROMO1 in early lesions. For the detection of early-stage cervical carcinoma and high-grade precancerous lesions, ROMO1 may be a useful auxiliary biomarker. Full article
Show Figures

Figure 1

18 pages, 14998 KB  
Communication
Nodal Marginal Zone Lymphoma with Prominent Expansion of PD-1+ T-Follicular Helper Cells: A Persistent Diagnostic Challenge with a Heterogeneous Mutational Architecture
by Stefania Crisci, Annarosaria De Chiara, Maria Oro, Maria Rivieccio, Annalisa Altobelli, Sara Mele, Letizia Sirica, Daniela Donnarumma, Matteo Bonanni, Annarosa Cuccaro, Alberto Fresa, Rosaria De Filippi and Antonio Pinto
Int. J. Mol. Sci. 2026, 27(1), 51; https://doi.org/10.3390/ijms27010051 - 20 Dec 2025
Viewed by 156
Abstract
Nodal marginal zone lymphoma (NMZL) is an indolent B-cell lymphoma that may pose diagnostic challenges due to the absence of distinct markers. In rare atypical cases, an overabundance of PD1+ T follicular helper (TFH) cells in tumor tissue may mimic peripheral T-cell lymphoma [...] Read more.
Nodal marginal zone lymphoma (NMZL) is an indolent B-cell lymphoma that may pose diagnostic challenges due to the absence of distinct markers. In rare atypical cases, an overabundance of PD1+ T follicular helper (TFH) cells in tumor tissue may mimic peripheral T-cell lymphoma (PTCL) of TFH origin, further complicating the diagnosis. A 72-year-old woman with progressive lymphadenopathy had a cervical lymph node biopsy showing a disrupted architecture with monomorphic nodules of CD20+/MNDA+ B-cells and a prominent central population of proliferating CD4+/PD1+ T-cells, initially suggestive of a PTCL-TFH. The bone marrow contained aggregates of CD20+ B-cells intermixed with CD3+/CD4+/PD1+ T-cells. Next-generation sequencing (NGS) revealed clonal immunoglobulin heavy-chain rearrangements in the lymph node and bone marrow, with T-cell receptor genes displaying a polyclonal pattern. Targeted NGS showed no PTCL-related alterations but identified NMZL-associated mutations with different distributions across lymph node and bone marrow compartments. NOTCH2 mutations (c.6418C>T; p.Gln2140*) were found in both tissues, while the (c.69+2T>A; p.?) TNFRSF14 gene mutation was only detected in the lymph node. The KMT2D gene displayed a frameshift variant in the lymph node (c.4801_4802delinsT; p.Arg1601Leufs*3) and an in-frame deletion (c.11756_11758del; p.Gln3919del) in the bone marrow. Notably, NGS and digital droplet PCR confirmed a TP53 frameshift mutation (c.902del; p.Pro301Glnfs*44) with a fractional abundance of 0.31% in the lymph node and a (c.742C>T; p.Arg248Trp) mutation (0.309%) in the bone marrow. Results underscore the importance of NGS-based clonality to diagnose NMZL with prominent PD1+ T-cell hyperplasia, and prompt further investigation into tissue-specific mutational signatures in these unusual cases. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

29 pages, 1473 KB  
Article
Global Dynamics of a Dual-Target HIV Model with Time Delays and Treatment Implications
by Hanan H. Almuashi and Miled El Hajji
Mathematics 2026, 14(1), 6; https://doi.org/10.3390/math14010006 - 19 Dec 2025
Cited by 1 | Viewed by 136
Abstract
We present a comprehensive mathematical analysis of a within-host dual-target HIV dynamics model, which explicitly incorporates the virus’s interactions with its two primary cellular targets: CD4+ T cells and macrophages. The model is formulated as a system of five nonlinear delay differential [...] Read more.
We present a comprehensive mathematical analysis of a within-host dual-target HIV dynamics model, which explicitly incorporates the virus’s interactions with its two primary cellular targets: CD4+ T cells and macrophages. The model is formulated as a system of five nonlinear delay differential equations, integrating three distinct discrete time delays to account for critical intracellular processes such as the development of productively infected cells and the maturation of new virions. We first establish the model’s biological well-posedness by proving the non-negativity and boundedness of solutions, ensuring all trajectories remain within a feasible region. The basic reproduction number, R0d, is derived using the next-generation matrix method and serves as a sharp threshold for disease dynamics. Analytical results demonstrate that the infection-free equilibrium is globally asymptotically stable (GAS) when R0d1, guaranteeing viral eradication from any initial state. Conversely, when R0d>1, a unique endemic equilibrium emerges and is proven to be GAS, representing a state of chronic infection. These global stability properties are rigorously established for both the non-delayed and delayed systems using carefully constructed Lyapunov functions and functionals, coupled with LaSalle’s invariance principle. A sensitivity analysis identifies viral production rates (p1,p2) and infection rates (β1,β2) as the most influential parameters on R0d, while the viral clearance rate (m) and maturation delay (τ3) have a suppressive effect. The model is extended to evaluate antiretroviral therapy (ART), revealing a critical treatment efficacy threshold ϵcr required to suppress the virus. Numerical simulations validate all theoretical findings and further investigate the dynamics under varying treatment efficacies and maturation delays, highlighting how these factors can shift the system from persistence to clearance. This study provides a rigorous mathematical framework for understanding HIV dynamics, with actionable insights for designing targeted treatment protocols aimed at achieving viral suppression. Full article
(This article belongs to the Special Issue Complex System Dynamics and Mathematical Biology)
Show Figures

Figure 1

19 pages, 5105 KB  
Article
Low Levels of Mouse γδ T Cell Development Persist in the Presence of Null Mutants of the LAT Adaptor
by Mikel M. Arbulo-Echevarria, Luis M. Fernandez-Aguilar, Elke Kurz, Inmaculada Vico-Barranco, Raquel Muñoz-Fernández, Isaac Narbona-Sánchez, Manuel Carrasco, Bernard Malissen, Michael L. Dustin and Enrique Aguado
Int. J. Mol. Sci. 2025, 26(24), 12186; https://doi.org/10.3390/ijms262412186 - 18 Dec 2025
Viewed by 140
Abstract
Activation through the T cell receptor (TCR) initiates a signaling cascade in T cells that induces extensive molecular and cellular changes. The adaptor protein Linker for Activation of T cells (LAT) plays an essential role in transducing activation and regulatory signals downstream of [...] Read more.
Activation through the T cell receptor (TCR) initiates a signaling cascade in T cells that induces extensive molecular and cellular changes. The adaptor protein Linker for Activation of T cells (LAT) plays an essential role in transducing activation and regulatory signals downstream of the TCR. Phosphorylation of LAT tyrosine residues recruits multiple signaling proteins, leading to the assembly of the LAT signalosome, which is crucial for relaying signals that regulate T cell development and function. We previously showed that substitution of a negatively charged amino acid segment preceding the fifth tyrosine residue of LAT (Tyr127 in humans or Tyr132 in mouse LAT) enhances some early TCR signaling events, whereas downstream responses, such as Ca2+ influx and Erk phosphorylation, are partially inhibited. To investigate the physiological relevance of this segment in vivo, we generated a new LAT knock-in mouse strain (LatNIL) in which the negatively charged segment was replaced with a non-charged sequence. Unexpectedly, this mutation led to an alternative splicing event in the Lat gene that excluded exons 6 and 7, resulting in a frameshift, a premature stop codon at residue 145, and the loss of the six C-terminal tyrosine residues of LAT. Homozygous LatNIL/NIL mice showed a phenotype similar to that of LAT-knockout and Lat4YF mice (in which the four C-terminal tyrosines had been mutated to phenylalanine). Interestingly, homozygous LatNIL/NIL mice exhibited a distinct population of γδ T cells in lymphoid organs, which has not been observed in LAT-KO or Lat4YF mice. These γδ T cells expressed higher levels of CD27 compared to those in wild-type and LAT-KO mice, suggesting altered activation or differentiation states. Together, these data highlight how subtle alterations in LAT structure can profoundly impact T cell signaling and lineage composition. Full article
(This article belongs to the Section Molecular Immunology)
Show Figures

Figure 1

30 pages, 1002 KB  
Review
Immunopathogenic Mechanisms in Connective Tissue Disease-Associated Interstitial Lung Disease: Incessant Loop of Immunity to Fibrosis
by Jae Ha Lee, Ji Hoon Jang, Sunggun Lee and Minyoung Her
Int. J. Mol. Sci. 2025, 26(24), 12126; https://doi.org/10.3390/ijms262412126 - 17 Dec 2025
Viewed by 303
Abstract
Connective tissue disease-associated interstitial lung disease (CTD-ILD) represents a significant cause of morbidity and mortality. It is characterized by the progressive convergence of chronic inflammation, immune dysregulation, and fibrotic remodeling in the lung parenchyma. While often conceptualized through a model of idiopathic pulmonary [...] Read more.
Connective tissue disease-associated interstitial lung disease (CTD-ILD) represents a significant cause of morbidity and mortality. It is characterized by the progressive convergence of chronic inflammation, immune dysregulation, and fibrotic remodeling in the lung parenchyma. While often conceptualized through a model of idiopathic pulmonary fibrosis (IPF), CTD-ILD is fundamentally an immune-driven pathology with distinct inflammatory mechanisms in which adaptive immunity plays a profound role. This narrative review explores the “inflammation–immunity–fibrosis continuum” in CTD-ILD, elaborating the intricate cellular and molecular pathways that distinguish it from IPF. We highlight the central role of persistent T-cell responses and B-cell dysregulation, which often occur within organized tertiary lymphoid structures in the lung. This review examines how these immune processes are propagated by multiple cytokine pathways, including the TGF-β/SMAD, JAK/STAT, and phosphodiesterase-4 signaling pathways, which serve as crucial links between inflammation and fibrosis. This distinct immune mechanism in CTD-ILD explains why immunomodulatory agents are a cornerstone of CTD-ILD treatment, in contrast to their limited efficacy in IPF, and emphasizes the current paradigm of combining immunosuppression with antifibrotic drugs to target the dual drivers of the disease. Full article
(This article belongs to the Special Issue New Advances in Autoimmune Diseases)
Show Figures

Figure 1

27 pages, 3096 KB  
Article
B Cells Can Trigger the T-Cell-Mediated Autoimmune Response Against Melanocytes in Psoriasis
by Mengwen He, Melissa Bernhardt, Akiko Arakawa, Song-Min Kim, Sigrid Vollmer, Burkard Summer, Yukiyasu Arakawa, Tatsushi Ishimoto, Andreas Schlosser and Jörg Christoph Prinz
Cells 2025, 14(24), 2002; https://doi.org/10.3390/cells14242002 - 16 Dec 2025
Viewed by 374
Abstract
Psoriasis vulgaris is a T-cell-mediated skin disease that may involve an autoimmune response against melanocytes. It develops through still unexplained pathomechanisms. Streptococcal tonsillopharyngitis is a major trigger of psoriasis onset and relapses. HLA-C*06:02 is the main psoriasis risk gene. Here we find that [...] Read more.
Psoriasis vulgaris is a T-cell-mediated skin disease that may involve an autoimmune response against melanocytes. It develops through still unexplained pathomechanisms. Streptococcal tonsillopharyngitis is a major trigger of psoriasis onset and relapses. HLA-C*06:02 is the main psoriasis risk gene. Here we find that B cells isolated from streptococci-infected tonsils or peripheral blood of HLA-C*06:02+ psoriasis patients stimulate an HLA-C*06:02-restricted melanocyte-reactive Vα3S1/Vβ13S1 T-cell receptor (TCR) from a lesional psoriatic CD8+ T cell clone in an IFN-γ-enhanced manner. Patients’ B cells furthermore induce proliferation of autologous blood CD8+ T cells. We identify several HLA-C*06:02-presented self-peptides in the immunopeptidomes we had isolated from four HLA-C*06:02 homozygous B-cell lines that stimulate the Vα3S1/Vβ13S1 TCR and differ from the melanocyte autoantigen recognized by this TCR. These data suggest that the proinflammatory environment of streptococcal tonsillopharyngitis may enable B cells to activate autoreactive CD8+ T cells that, owing to the polyspecificity of T-cell receptors, recognize several B-cell self-peptides presented by HLA-C*06:02 and subsequently cross-react against melanocytes in the skin, thereby triggering psoriasis. The capacity of B cells to stimulate a cross-reactive autoimmune response through HLA class I-presented B-cell peptides is a previously unknown mechanism in the induction of autoimmunity that could explain psoriasis onset and persistence. Full article
(This article belongs to the Special Issue Genetic and Cellular Basis of Autoimmune Diseases)
Show Figures

Graphical abstract

24 pages, 6497 KB  
Article
Curcumin Modulates TIGIT/Neuropilin-1 to Regulate T-Cell Immune Homeostasis in Ulcerative Colitis
by Yazhen Liu, Jiaqi Huang, Ji Yu, Luxin Fu, Ronglong Huang, Jing Liu, Bailin Deng, You-Bao Zhong, Duanyong Liu and Haimei Zhao
Foods 2025, 14(24), 4323; https://doi.org/10.3390/foods14244323 - 15 Dec 2025
Viewed by 322
Abstract
(1) Background: Ulcerative colitis (UC) is a persistent inflammatory condition of the intestine, characterized by dysregulated T cell-mediated immune responses. Curcumin (CUR), a common food additive and health supplement, is noted for possessing anti-inflammatory and immunomodulatory properties. Nevertheless, the molecular mechanisms underlying its [...] Read more.
(1) Background: Ulcerative colitis (UC) is a persistent inflammatory condition of the intestine, characterized by dysregulated T cell-mediated immune responses. Curcumin (CUR), a common food additive and health supplement, is noted for possessing anti-inflammatory and immunomodulatory properties. Nevertheless, the molecular mechanisms underlying its therapeutic effects remain incompletely elucidated. This research aims to investigate the therapeutic mechanisms of CUR in UC, focusing on its role in restoring T cell homeostasis by modulating TIGIT and Neuropilin-1 (NRP1). (2) Methods: We employed a DSS-induced murine colitis model, combined with network pharmacology, molecular docking, protein–protein interaction docking, molecular dynamics simulations, and invitro assays with Jurkat T cells. (3) Results: CUR markedly ameliorated clinical manifestations and histopathology in DSS-treated mice, restoring the balance of T cell and memory T cell subsets. Computational predictions and experimental validation showed that CUR downregulated TIGIT and NRP1 expression in inflamed colonic tissue and directly inhibited their expression in activated T cells invitro. (4) Conclusions: This study reveals a novel immunoregulatory mechanism of this natural compound. These findings suggest CUR modulates TIGIT/NRP1 to inhibit excessive T cell activation and restore immune homeostasis in UC. Full article
(This article belongs to the Section Food Nutrition)
Show Figures

Graphical abstract

Back to TopTop