Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 29.611
1.
Yakugaku Zasshi ; 144(5): 475-481, 2024.
Article Ja | MEDLINE | ID: mdl-38692920

Zinc is one of the essential trace elements, and is involved in various functions in the body. Zinc deficiency is known to cause immune abnormalities, but the mechanism is not fully understood. Therefore, we focused our research on tumor immunity to elucidate the effect of zinc on colorectal cancer and its mechanisms. Mice were treated with azoxymethane (AOM) and dextran sodium sulfate (DSS) to develop colorectal cancer, then the relationship between zinc content in the diet and the number and area of tumors in the colon was observed. The number of tumors in the colon was significantly higher in the no-zinc-added diet group compared to the normal zinc intake group, and about half the number in the high-zinc-intake group compared to the normal-zinc-intake group. In T-cell-deficient mice, the number of tumors in the high-zinc-intake group was similar to that in the normal-zinc-intake group, suggesting that the inhibitory effect of zinc was dependent on T cells. Furthermore, we found that the amount of granzyme B transcript released by cytotoxic T cells upon antigen stimulation was significantly increased by the addition of zinc. We also showed that granzyme B transcriptional activation by zinc addition was dependent on calcineurin activity. Collectively, we have shown that zinc exerts its tumor-suppressive effect by acting on cytotoxic T cells, the center of cellular immunity, and that it increases the transcription of granzyme B, one of the key molecules involved in tumor immunity. In this symposium, we would like to introduce our latest data on the relationship between zinc and tumor immunity.


Colorectal Neoplasms , Immunity, Cellular , Zinc , Animals , Colorectal Neoplasms/immunology , Colorectal Neoplasms/etiology , Colorectal Neoplasms/prevention & control , Mice , Humans , Granzymes/metabolism , T-Lymphocytes, Cytotoxic/immunology , Azoxymethane , Disease Models, Animal
2.
J Math Biol ; 89(1): 6, 2024 May 19.
Article En | MEDLINE | ID: mdl-38762831

Multiple infections enable the recombination of different strains, which may contribute to viral diversity. How multiple infections affect the competition dynamics between the two types of strains, the wild and the immune escape mutant, remains poorly understood. This study develops a novel mathematical model that includes the two strains, two modes of viral infection, and multiple infections. For the representative double-infection case, the reproductive numbers are derived and global stabilities of equilibria are obtained via the Lyapunov direct method and theory of limiting systems. Numerical simulations indicate similar viral dynamics regardless of multiplicities of infections though the competition between the two strains would be the fiercest in the case of quadruple infections. Through sensitivity analysis, we evaluate the effect of parameters on the set-point viral loads in the presence and absence of multiple infections. The model with multiple infections predict that there exists a threshold for cytotoxic T lymphocytes (CTLs) to minimize the overall viral load. Weak or strong CTLs immune response can result in high overall viral load. If the strength of CTLs maintains at an intermediate level, the fitness cost of the mutant is likely to have a significant impact on the evolutionary dynamics of mutant viruses. We further investigate how multiple infections alter the viral dynamics during the combination antiretroviral therapy (cART). The results show that viral loads may be underestimated during cART if multiple-infection is not taken into account.


Computer Simulation , HIV Infections , Immune Evasion , Mathematical Concepts , Models, Biological , T-Lymphocytes, Cytotoxic , Viral Load , Humans , HIV Infections/immunology , HIV Infections/virology , HIV Infections/drug therapy , T-Lymphocytes, Cytotoxic/immunology , Immune Evasion/immunology , Coinfection/immunology , Coinfection/virology , HIV-1/immunology , HIV-1/genetics , Basic Reproduction Number/statistics & numerical data , Models, Immunological , Mutation
3.
Sci Rep ; 14(1): 10842, 2024 05 12.
Article En | MEDLINE | ID: mdl-38735993

Yellow fever outbreaks are prevalent, particularly in endemic regions. Given the lack of an established treatment for this disease, significant attention has been directed toward managing this arbovirus. In response, we developed a multiepitope vaccine designed to elicit an immune response, utilizing advanced immunoinformatic and molecular modeling techniques. To achieve this, we predicted B- and T-cell epitopes using the sequences from all structural (E, prM, and C) and nonstructural proteins of 196 YFV strains. Through comprehensive analysis, we identified 10 cytotoxic T-lymphocyte (CTL) and 5T-helper (Th) epitopes that exhibited overlap with B-lymphocyte epitopes. These epitopes were further evaluated for their affinity to a wide range of human leukocyte antigen system alleles and were rigorously tested for antigenicity, immunogenicity, allergenicity, toxicity, and conservation. These epitopes were linked to an adjuvant ( ß -defensin) and to each other using ligands, resulting in a vaccine sequence with appropriate physicochemical properties. The 3D structure of this sequence was created, improved, and quality checked; then it was anchored to the Toll-like receptor. Molecular Dynamics and Quantum Mechanics/Molecular Mechanics simulations were employed to enhance the accuracy of docking calculations, with the QM portion of the simulations carried out utilizing the density functional theory formalism. Moreover, the inoculation model was able to provide an optimal codon sequence that was inserted into the pET-28a( +) vector for in silico cloning and could even stimulate highly relevant humoral and cellular immunological responses. Overall, these results suggest that the designed multi-epitope vaccine can serve as prophylaxis against the yellow fever virus.


Epitopes, T-Lymphocyte , Yellow Fever Vaccine , Yellow Fever , Yellow fever virus , Yellow Fever Vaccine/immunology , Yellow fever virus/immunology , Yellow fever virus/genetics , Humans , Yellow Fever/prevention & control , Yellow Fever/immunology , Epitopes, T-Lymphocyte/immunology , Epitopes, B-Lymphocyte/immunology , Vaccinology/methods , Models, Molecular , Vaccine Development , Molecular Dynamics Simulation , T-Lymphocytes, Cytotoxic/immunology
4.
Biochem Biophys Res Commun ; 718: 150058, 2024 Jul 23.
Article En | MEDLINE | ID: mdl-38729076

The therapeutic efficacy of radiotherapy (RT) is primarily driven by two factors: biophysical DNA damage in cancer cells and radiation-induced anti-tumor immunity. However, Anti-tumor immune responses between X-ray RT (XRT) and carbon-ion RT (CIRT) remain unclear. In this study, we, employed mouse models to assess the immunological contribution, especially cytotoxic T-lymphocyte (CTL)-mediated immunity, to the therapeutic effectiveness of XRT and CIRT in shrinking tumors. We irradiated mouse intradermal tumors of B16F10-ovalbumin (OVA) mouse melanoma cells and 3LL-OVA mouse lung cancer cells with carbon-ion beams or X-rays in the presence or absence of CTLs. CTL removal was performed by administration of anti-CD8 monoclonal antibody (mAb) in mice. Based on tumor growth delay, we determined the tumor growth and regression curves. The enhancement ratio (ER) of the slope of regression lines in the presence of CTLs, relative to the absence of CTLs, indicates the dependency of RT on CTLs for shrinking mouse tumors, and the biological effectiveness (RBE) of CIRT relative to XRT were calculated. Tumor growth curves revealed that the elimination of CD8+ CTLs by administrating anti-CD8 mAb accelerated tumor growth compared to the presence of CTLs in both RTs. The ERs were larger in CIRT compared to XRT in the B16F10-OVA tumor models, but not in the 3LL-OVA models, suggesting a greater contribution of CTL-mediated anti-tumor immunity to tumor reduction in CIRT compared to XRT in the B16F10-OVA tumor model. In addition, the RBE values for both models were larger in the presence of CTLs compared to models without CTLs, suggesting that CIRT may utilize CTL-mediated anti-tumor immunity more than X-ray. The findings from this study suggest that although immunological contribution to therapeutic efficacy may vary depending on the type of tumor cell, CIRT utilizes CTL-mediated immunity to a greater extent compared to XRT.


Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic , Animals , T-Lymphocytes, Cytotoxic/immunology , Mice , Cell Line, Tumor , Melanoma, Experimental/immunology , Melanoma, Experimental/radiotherapy , Melanoma, Experimental/therapy , Melanoma, Experimental/pathology , Heavy Ion Radiotherapy/methods , X-Ray Therapy , Female , Lung Neoplasms/immunology , Lung Neoplasms/radiotherapy , Lung Neoplasms/therapy , Lung Neoplasms/pathology
5.
ACS Nano ; 18(20): 13226-13240, 2024 May 21.
Article En | MEDLINE | ID: mdl-38712706

Oncogene activation and epigenome dysregulation drive tumor initiation and progression, contributing to tumor immune evasion and compromising the clinical response to immunotherapy. Epigenetic immunotherapy represents a promising paradigm in conquering cancer immunosuppression, whereas few relevant drug combination and delivery strategies emerge in the clinic. This study presents a well-designed triune nanomodulator, termed ROCA, which demonstrates robust capabilities in tumor epigenetic modulation and immune microenvironment reprogramming for cancer epigenetic immunotherapy. The nanomodulator is engineered from a nanoscale framework with epigenetic modulation and cascaded catalytic activity, which self-assembles into a nanoaggregate with tumor targeting polypeptide decoration that enables loading of the immunogenic cell death (ICD)-inducing agent. The nanomodulator releases active factors specifically triggered in the tumor microenvironment, represses oncogene expression, and initiates the type 1 T helper (TH1) cell chemokine axis by reversing DNA hypermethylation. This process, together with ICD induction, fundamentally reprograms the tumor microenvironment and significantly enhances the rejuvenation of exhausted cytotoxic T lymphocytes (CTLs, CD8+ T cells), which synergizes with the anti-PD-L1 immune checkpoint blockade and results in a boosted antitumor immune response. Furthermore, this strategy establishes long-term immune memory and effectively prevents orthotopic colon cancer relapse. Therefore, the nanomodulator holds promise as a standalone epigenetic immunotherapy agent or as part of a combination therapy with immune checkpoint inhibitors in preclinical cancer models, broadening the array of combinatorial strategies in cancer immunotherapy.


Epigenesis, Genetic , Immunotherapy , T-Lymphocytes, Cytotoxic , Tumor Microenvironment , Animals , Epigenesis, Genetic/drug effects , Mice , T-Lymphocytes, Cytotoxic/immunology , Humans , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Nanoparticles/chemistry , Mice, Inbred C57BL , Neoplasms/therapy , Neoplasms/immunology
6.
Hum Vaccin Immunother ; 20(1): 2352908, 2024 Dec 31.
Article En | MEDLINE | ID: mdl-38780076

Cervical cancer, among the deadliest cancers affecting women globally, primarily arises from persistent infection with high-risk human papillomavirus (HPV). To effectively combat persistent infection and prevent the progression of precancerous lesions into malignancy, a therapeutic HPV vaccine is under development. This study utilized an immunoinformatics approach to predict epitopes of cytotoxic T lymphocytes (CTLs) and helper T lymphocytes (HTLs) using the E6 and E7 oncoproteins of the HPV16 strain as target antigens. Subsequently, through meticulous selection of T-cell epitopes and other necessary elements, a multi-epitope vaccine was constructed, exhibiting good immunogenic, physicochemical, and structural characteristics. Furthermore, in silico simulations showed that the vaccine not only interacted well with toll-like receptors (TLR2/TLR3/TLR4), but also induced a strong innate and adaptive immune response characterized by elevated Th1-type cytokines, such as interferon-gamma (IFN-γ) and interleukin-2 (IL2). Additionally, our study investigated the effects of different immunization intervals on immune responses, aiming to optimize a time-efficient immunization program. In animal model experiments, the vaccine exhibited robust immunogenic, therapeutic, and prophylactic effects. Administered thrice, it consistently induced the expansion of specific CD4 and CD8 T cells, resulting in substantial cytokines release and increased proliferation of memory T cell subsets in splenic cells. Overall, our findings support the potential of this multi-epitope vaccine in combating HPV16 infection and signify its candidacy for future HPV vaccine development.


Through the stringent selection of T-cell epitopes and other necessary elements, a novel multi-epitope vaccine targeting HPV 16 E6 and E7 oncoproteins was constructed using an immunoinformatics approach.The vaccine designed can induce both cellular and humoral immune responses, encompassing all the required immunogenic, physicochemical, and structural characteristics for an ideal vaccine design. Moreover, it offers decent worldwide coverage.In animal studies, the vaccine demonstrated strong immune responses, including expansion of CD4 and CD8 T cells, cytokine release, and enhanced memory T cell proliferation, resulting in long-term anti-tumor effects, inhibition of tumor growth, and prolonged survival in tumor-bearing mice.The immunological evaluation of the designed vaccine suggests its potential as a novel vaccine candidate against HPV 16.


Epitopes, T-Lymphocyte , Human papillomavirus 16 , Oncogene Proteins, Viral , Papillomavirus Infections , Papillomavirus Vaccines , Uterine Cervical Neoplasms , Vaccines, DNA , Female , Papillomavirus Vaccines/immunology , Papillomavirus Vaccines/administration & dosage , Human papillomavirus 16/immunology , Vaccines, DNA/immunology , Vaccines, DNA/administration & dosage , Papillomavirus Infections/prevention & control , Papillomavirus Infections/immunology , Epitopes, T-Lymphocyte/immunology , Animals , Oncogene Proteins, Viral/immunology , Oncogene Proteins, Viral/genetics , Uterine Cervical Neoplasms/prevention & control , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/virology , Papillomavirus E7 Proteins/immunology , Mice , Humans , T-Lymphocytes, Cytotoxic/immunology , Repressor Proteins/immunology , T-Lymphocytes, Helper-Inducer/immunology , Mice, Inbred C57BL , Interferon-gamma/metabolism , Interferon-gamma/immunology
7.
Int J Biol Macromol ; 269(Pt 2): 132177, 2024 Jun.
Article En | MEDLINE | ID: mdl-38729484

Tumor vaccine, which can effectively prevent tumor recurrence and metastasis, is a promising tool in tumor immunotherapy. However, heterogeneity of tumors and the inability to achieve a cascade effect limit the therapeutic effects of most developing tumor vaccine. We have developed a cascading immunoinducible in-situ mannose-functionalized polydopamine loaded with imiquimod phenylboronic hyaluronic acid nanocomposite gel vaccine (M/P-PDA@IQ PHA) through a boronic ester-based reaction. This reaction utilizes mannose-functionalized polydopamine loaded with imiquimod (M/P-PDA@IQ NAs) as a cross-linking agent to react with phenylboronic-grafted hyaluronic acid. Under near-infrared light irradiation, the M/P-PDA@IQ PHA caused local hyperthermia to trigger immunogenic cell death of tumor cells and tumor-associated antigens (TAAs) releasing. Subsequently, the M/P-PDA@IQ NAs which were gradually released by the pH/ROS/GSH-triggered degradation of M/P-PDA@IQ PHA, could capture and deliver these TAAs to lymph nodes. Finally, the M/P-PDA@IQ NAs facilitated maturation and cross-presentation of dendritic cells, as well as activation of cytotoxic T lymphocytes. Overall, the M/P-PDA@IQ PHA could serve as a novel in situ vaccine to stimulate several key nodes including TAAs release and capture, targeting lymph nodes and enhanced dendritic cells uptake and maturation as well as T cells activation. This cascading immune activation strategy can effectively elicit antitumor immune response.


Cancer Vaccines , Hyaluronic Acid , Hydrogels , Indoles , Nanoparticles , Polymers , Hyaluronic Acid/chemistry , Polymers/chemistry , Cancer Vaccines/chemistry , Cancer Vaccines/immunology , Indoles/chemistry , Indoles/pharmacology , Animals , Mice , Hydrogels/chemistry , Nanoparticles/chemistry , Humans , Imiquimod/chemistry , Imiquimod/pharmacology , Dendritic Cells/immunology , Vaccination , Cell Line, Tumor , Immunotherapy/methods , Cross-Linking Reagents/chemistry , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/drug effects
8.
Front Immunol ; 15: 1338218, 2024.
Article En | MEDLINE | ID: mdl-38742109

Cytotoxic T lymphocyte (CTL) motility is an important feature of effective CTL responses and is impaired when CTLs become exhausted, e.g. during chronic retroviral infections. A prominent T cell exhaustion marker is programmed cell death protein 1 (PD-1) and antibodies against the interaction of PD-1 and PD-ligand 1 (PD-L1) are known to improve CTL functions. However, antibody blockade affects all PD-1/PD-L1-expressing cell types, thus, the observed effects cannot be attributed selectively to CTLs. To overcome this problem, we performed CRISPR/Cas9 based knockout of the PD-1 coding gene PDCD1 in naïve Friend Retrovirus (FV)-specific CTLs. We transferred 1,000 of these cells into mice where they proliferated upon FV-infection. Using intravital two-photon microscopy we visualized CTL motility in the bone marrow and evaluated cytotoxic molecule expression by flow cytometry. Knockout of PDCD1 improved the CTL motility at 14 days post infection and enhanced the expression of cytotoxicity markers. Our data show the potential of genetic tuning of naive antiviral CTLs and might be relevant for future designs of improved T cell-mediated therapies.


Cell Movement , Mice, Knockout , Programmed Cell Death 1 Receptor , Retroviridae Infections , T-Lymphocytes, Cytotoxic , Animals , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/immunology , Mice , Cell Movement/genetics , Retroviridae Infections/immunology , T-Lymphocytes, Cytotoxic/immunology , Mice, Inbred C57BL , Friend murine leukemia virus/immunology , Gene Knockout Techniques , CD8-Positive T-Lymphocytes/immunology , CRISPR-Cas Systems , Cytotoxicity, Immunologic
9.
J Control Release ; 369: 556-572, 2024 May.
Article En | MEDLINE | ID: mdl-38580136

Vaccines represent one of the most powerful and cost-effective innovations for controlling a wide range of infectious diseases caused by various viruses and bacteria. Unlike mRNA and DNA-based vaccines, subunit vaccines carry no risk of insertional mutagenesis and can be lyophilized for convenient transportation and long-term storage. However, existing adjuvants are often associated with toxic effect and reactogenicity, necessitating expanding the repertoire of adjuvants with better biocompatibility, for instance, designing self-adjuvating polymeric carriers. We herein report a novel subunit vaccine delivery platform constructed via in situ free radical polymerization of C7A (2-(Hexamethyleneimino) ethyl methacrylate) and acrylamide around the surface of individual protein antigens. Using ovalbumin (OVA) as a model antigen, we observed substantial increases in both diameter (∼70 nm) and surface potential (-1.18 mV) following encapsulation, referred to as n(OVA)C7A. C7A's ultra pH sensitivity with a transition pH around 6.9 allows for rapid protonation in acidic environments. This property facilitates crucial processes such as endosomal escape and major histocompatibility complex (MHC)-I-mediated antigen presentation, culminating in the substantial CD8+ T cell activation. Additionally, compared to OVA nanocapsules without the C7A components and native OVA without modifications, we observed heightened B cell activation within the germinal center, along with remarkable increases in serum antibody and cytokine production. It's important to note that mounting evidence suggests that adjuvant effects, particularly its targeted stimulation of type I interferons (IFNs), can contribute to advantageous adaptive immune responses. Beyond its exceptional potency, the nanovaccine also demonstrated robust formation of immune memory and exhibited a favorable biosafety profile. These findings collectively underscore the promising potential of our nanovaccine in the realm of immunotherapy and vaccine development.


Mice, Inbred C57BL , Ovalbumin , T-Lymphocytes, Cytotoxic , Animals , Ovalbumin/immunology , Ovalbumin/administration & dosage , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/drug effects , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/chemistry , Female , Methacrylates/chemistry , Polymers/chemistry , Polymers/administration & dosage , Nanoparticles/chemistry , Nanoparticles/administration & dosage , Mice , Vaccines/administration & dosage , Vaccines/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Nanovaccines
10.
Neuromolecular Med ; 26(1): 17, 2024 Apr 29.
Article En | MEDLINE | ID: mdl-38684592

Post-stroke neuroinflammation affects the damage and recovery of neurological functions. T cells including CD8+ T cells were present in the ipsilateral hemisphere in the subacute and late phases of ischemic stroke. However, the potential roles of CD8+ T cell subsets in the progression of neuroinflammation have not been characterized. In the current mouse transient middle cerebral artery occlusion model, we investigated the existence of CD8+ T cell subsets in the ipsilateral hemisphere in the subacute and late phases of stroke. We found that ipsilateral CD8+ T cells were present on post-stroke day 3 and increased on post-stroke day 30. The day-3 ipsilateral CD8+ T cells predominantly produced interferon-γ (IFN-γ), while the day-30 ipsilateral CD8+ T cells co-expressed IFN-γ and interleukin-17A (IL-17A). In addition, evaluation of cytokines and transcription factors of the day-30 ipsilateral CD8+ T cells revealed the presence of T cytotoxic 1 (Tc1), T cytotoxic 17 (Tc17), and T cytotoxic 17/1 (Tc17/1) cells. Furthermore, based on the expression of a series of chemokine/cytokine receptors, viable ipsilateral Tc1, Tc17, and Tc17.1 cells were identified and enriched from the day-30 ipsilateral CD8+ T cells, respectively. Co-culture of microglia with ipsilateral Tc1, Tc17, or Tc17.1 cells indicated that the three CD8+ T cell subsets up-regulated the expression of pro-inflammatory mediators by microglia, with Tc17.1 cells being the most potent cell in doing so. Collectively, this study sheds light on the contributions of Tc1, Tc17, and Tc17.1 cells to long-term neuroinflammation after ischemic stroke.


Infarction, Middle Cerebral Artery , Interleukin-17 , Mice, Inbred C57BL , Microglia , Neuroinflammatory Diseases , T-Lymphocytes, Cytotoxic , Animals , Microglia/metabolism , Mice , Male , Infarction, Middle Cerebral Artery/immunology , Infarction, Middle Cerebral Artery/pathology , T-Lymphocytes, Cytotoxic/immunology , Neuroinflammatory Diseases/etiology , Ischemic Stroke/immunology , Interferon-gamma/biosynthesis , Brain , Th17 Cells/immunology , Disease Models, Animal , CD8-Positive T-Lymphocytes , Coculture Techniques , Cells, Cultured
11.
Cancer Lett ; 591: 216893, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38636892

The oncogenic properties of Nucleobindin2 (NUCB2) have been observed in various cancer types. Nevertheless, the precise understanding of the biological functions and regulatory mechanisms of NUCB2 in osteosarcoma remains limited. This investigation reported that NUCB2 was significantly increased upon glucose deprivation-induced metabolic stress. Elevated NUCB2 suppressed glucose deprivation-induced cell death and reactive oxygen species (ROS) increase. Depletion of NUCB2 resulted in a reduction in osteosarcoma cell proliferation as well as metastatic potential in vitro and in vivo. Mechanically, NUCB2 ablation suppressed C-X-C Motif Chemokine Ligand 8 (CXCL8) expression which then reduced programmed cell death 1 ligand 1 (PD-L1) expression and stimulated anti-tumor immunity mediated through cytotoxic T cells. Importantly, a combination of NUCB2 depletion with anti-PD-L1 treatment improved anti-tumor T-cell immunity in vivo. Moreover, we further demonstrated that NUCB2 interacted with NUCKS1 to inhibit its degradation, which is responsible for the transcriptional regulation of CXCL8 expression. Altogether, the outcome emphasizes the function of NUCB2 in osteosarcoma and indicates that NUCB2 elevates osteosarcoma progression and immunosuppressive microenvironment through the NUCKS1/CXCL8 pathway.


Bone Neoplasms , Calcium-Binding Proteins , Disease Progression , Interleukin-8 , Osteosarcoma , Tumor Microenvironment , Osteosarcoma/immunology , Osteosarcoma/pathology , Osteosarcoma/metabolism , Osteosarcoma/genetics , Humans , Animals , Cell Line, Tumor , Interleukin-8/metabolism , Interleukin-8/genetics , Mice , Bone Neoplasms/immunology , Bone Neoplasms/pathology , Bone Neoplasms/metabolism , Bone Neoplasms/genetics , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/genetics , Tumor Microenvironment/immunology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Cell Proliferation , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Gene Expression Regulation, Neoplastic , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , T-Lymphocytes, Cytotoxic/immunology , Signal Transduction , Reactive Oxygen Species/metabolism
12.
Neurol Neuroimmunol Neuroinflamm ; 11(4): e200250, 2024 Jul.
Article En | MEDLINE | ID: mdl-38662990

BACKGROUND AND OBJECTIVES: The role of B cells in the pathogenic events leading to relapsing multiple sclerosis (R-MS) has only been recently elucidated. A pivotal step in defining this role has been provided by therapeutic efficacy of anti-CD20 monoclonal antibodies. Indeed, treatment with anti-CD20 can also alter number and function of other immune cells not directly expressing CD20 on their cell surface, whose activities can contribute to unknown aspects influencing therapeutic efficacy. We examined the phenotype and function of cytotoxic lymphocytes and Epstein-Barr virus (EBV)-specific immune responses in people with R-MS before and after ocrelizumab treatment. METHODS: In this prospective study, we collected blood samples from people with R-MS (n = 41) before and 6 and 12 months after initiating ocrelizumab to assess the immune phenotype and the indirect impact on cytotoxic functions of CD8+ T and NK cells. In addition, we evaluated the specific anti-EBV proliferative responses of both CD8+ T and NK lymphocytes as surrogate markers of anti-EBV activity. RESULTS: We observed that while ocrelizumab depleted circulating B cells, it also reduced the expression of activation and migratory markers on both CD8+ T and NK cells as well as their in vitro cytotoxic activity. A comparable pattern in the modulation of immune molecules by ocrelizumab was observed in cytotoxic cells even when patients with R-MS were divided into groups based on their prior disease-modifying treatment. These effects were accompanied by a significant and selective reduction of CD8+ T-cell proliferation in response to EBV antigenic peptides. DISCUSSION: Taken together, our findings suggest that ocrelizumab-while depleting B cells-affects the cytotoxic function of CD8+ and NK cells, whose reduced cross-activity against myelin antigens might also contribute to its therapeutic efficacy during MS.


Antibodies, Monoclonal, Humanized , CD8-Positive T-Lymphocytes , Herpesvirus 4, Human , Immunologic Factors , Humans , Antibodies, Monoclonal, Humanized/pharmacology , Female , Adult , Male , Herpesvirus 4, Human/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Middle Aged , Immunologic Factors/pharmacology , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Multiple Sclerosis, Relapsing-Remitting/immunology , Multiple Sclerosis, Relapsing-Remitting/blood , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Prospective Studies , Cell Proliferation/drug effects , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Lymphocyte Activation/drug effects
13.
J Math Biol ; 88(6): 75, 2024 Apr 30.
Article En | MEDLINE | ID: mdl-38689137

The aim of this paper is to develop and investigate a novel mathematical model of the dynamical behaviors of chronic hepatitis B virus infection. The model includes exposed infected hepatocytes, intracellular HBV DNA-containing capsids, uses a general incidence function for viral infection covering a variety of special cases available in the literature, and describes the interaction of cytotoxic T lymphocytes that kill the infected hepatocytes and the magnitude of B-cells that send antibody immune defense to neutralize free virions. Further, one time delay is incorporated to account for actual capsids production. The other time delays are used to account for maturation of capsids and free viruses. We start with the analysis of the proposed model by establishing the local and global existence, uniqueness, non-negativity and boundedness of solutions. After defined the threshold parameters, we discuss the stability properties of all possible steady state constants by using the crafty Lyapunov functionals, the LaSalle's invariance principle and linearization methods. The impacts of the three time delays on the HBV infection transmission are discussed through local and global sensitivity analysis of the basic reproduction number and of the classes of infected states. Finally, an application is provided and numerical simulations are performed to illustrate and interpret the theoretical results obtained. It is suggested that, a good strategy to eradicate or to control HBV infection within a host should concentrate on any drugs that may prolong the values of the three delays.


Adaptive Immunity , Capsid , Computer Simulation , Hepatitis B virus , Hepatitis B, Chronic , Hepatocytes , Mathematical Concepts , Hepatocytes/immunology , Hepatocytes/virology , Hepatitis B virus/immunology , Humans , Capsid/immunology , Adaptive Immunity/immunology , Hepatitis B, Chronic/immunology , Hepatitis B, Chronic/virology , Hepatitis B, Chronic/transmission , Models, Immunological , T-Lymphocytes, Cytotoxic/immunology , Basic Reproduction Number/statistics & numerical data , B-Lymphocytes/immunology , DNA, Viral/immunology , Models, Biological
14.
Cell Rep Med ; 5(5): 101505, 2024 May 21.
Article En | MEDLINE | ID: mdl-38614095

Immune checkpoint inhibitors (ICIs) represent a promising treatment for hepatocellular carcinoma (HCC) due to their capacity for abundant lymphocyte infiltration. However, some patients with HCC respond poorly to ICI therapy due to the presence of various immunosuppressive factors in the tumor microenvironment. Our research reveals that a macrophage-coated tumor cluster (MCTC) signifies a unique spatial structural organization in HCC correlating with diminished recurrence-free survival and overall survival in a total of 572 HCC cases from 3 internal cohorts and 2 independent external validation cohorts. Mechanistically, tumor-derived macrophage-associated lectin Mac-2 binding protein (M2BP) induces MCTC formation and traps immunocompetent cells at the edge of MCTCs to induce intratumoral cytotoxic T cell exclusion and local immune deprivation. Blocking M2BP with a Mac-2 antagonist might provide an effective approach to prevent MCTC formation, enhance T cell infiltration, and thereby improve the efficacy of ICI therapy in HCC.


Carcinoma, Hepatocellular , Immunotherapy , Liver Neoplasms , Macrophages , Tumor Microenvironment , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Humans , Macrophages/immunology , Immunotherapy/methods , Animals , Tumor Microenvironment/immunology , Mice , Drug Resistance, Neoplasm/drug effects , Male , Female , Cell Line, Tumor , Neoplasm Invasiveness , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Middle Aged , T-Lymphocytes, Cytotoxic/immunology , Tumor-Associated Macrophages/immunology
15.
Nat Immunol ; 25(5): 834-846, 2024 May.
Article En | MEDLINE | ID: mdl-38561495

Cancer remains one of the leading causes of mortality worldwide, leading to increased interest in utilizing immunotherapy strategies for better cancer treatments. In the past decade, CD103+ T cells have been associated with better clinical prognosis in patients with cancer. However, the specific immune mechanisms contributing toward CD103-mediated protective immunity remain unclear. Here, we show an unexpected and transient CD61 expression, which is paired with CD103 at the synaptic microclusters of T cells. CD61 colocalization with the T cell antigen receptor further modulates downstream T cell antigen receptor signaling, improving antitumor cytotoxicity and promoting physiological control of tumor growth. Clinically, the presence of CD61+ tumor-infiltrating T lymphocytes is associated with improved clinical outcomes, mediated through enhanced effector functions and phenotype with limited evidence of cellular exhaustion. In conclusion, this study identified an unconventional and transient CD61 expression and pairing with CD103 on human immune cells, which potentiates a new target for immune-based cellular therapies.


Antigens, CD , Apyrase , Integrin alpha Chains , Receptors, Antigen, T-Cell , Signal Transduction , Animals , Humans , Mice , Antigens, CD/metabolism , Antigens, CD/immunology , Cell Line, Tumor , Cytotoxicity, Immunologic , Integrin alpha Chains/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasms/immunology , Neoplasms/therapy , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , T-Lymphocytes, Cytotoxic/immunology
16.
Front Immunol ; 15: 1337973, 2024.
Article En | MEDLINE | ID: mdl-38665920

Cytotoxic T lymphocytes are the primary effector immune cells responsible for protection against cancer, as they target peptide neoantigens presented through the major histocompatibility complex (MHC) on cancer cells, leading to cell death. Targeting peptide-MHC (pMHC) complex offers a promising strategy for immunotherapy due to their specificity and effectiveness against cancer. In this work, we exploit the acidic tumor micro-environment to selectively deliver antigenic peptides to cancer using pH(low) insertion peptides (pHLIP). We demonstrated the delivery of MHC binding peptides directly to the cytoplasm of melanoma cells resulted in the presentation of antigenic peptides on MHC, and activation of T cells. This work highlights the potential of pHLIP as a vehicle for the targeted delivery of antigenic peptides and its presentation via MHC-bound complexes on cancer cell surface for activation of T cells with implications for enhancing anti-cancer immunotherapy.


Antigen Presentation , Membrane Proteins , Oligopeptides , Humans , Antigen Presentation/immunology , Animals , Antigens, Neoplasm/immunology , Cell Line, Tumor , Immunotherapy/methods , Acidosis/immunology , Lymphocyte Activation/immunology , Tumor Microenvironment/immunology , Mice , T-Lymphocytes, Cytotoxic/immunology , Peptides/immunology , Hydrogen-Ion Concentration , Melanoma/immunology , Melanoma/therapy
17.
Adv Sci (Weinh) ; 11(20): e2305832, 2024 May.
Article En | MEDLINE | ID: mdl-38564766

Effective presentation of antigens by dendritic cells (DC) is essential for achieving a robust cytotoxic T lymphocytes (CTLs) response, in which cDC1 is the key DC subtype for high-performance activation of CTLs. However, low cDC1 proportion, complex process, and high cost severely hindered cDC1 generation and application. Herein, the study proposes an in situ cDC1 recruitment and activation strategy with simultaneous inhibiting cancer stemness for inducing robust CTL responses and enhancing the anti-tumor effect. Fms-like tyrosine kinase 3 ligand (FLT3L), Poly I:C, and Nap-CUM (NCUM), playing the role of cDC1 recruitment, cDC1 activation, inducing antigen release and decreasing tumor cell stemness, respectively, are co-encapsulated in an in situ hydrogel vaccine (FP/NCUM-Gel). FP/NCUM-Gel is gelated in situ after intra-tumoral injection. With the near-infrared irradiation, tumor cell immunogenic cell death occurred, tumor antigens and immunogenic signals are released in situ. cDC1 is recruited to tumor tissue and activated for antigen cross-presentation, followed by migrating to lymph nodes and activating CTLs. Furthermore, tumor cell stemness are inhibited by napabucasin, which can help CTLs to achieve comprehensive tumor killing. Collectively, the proposed strategy of cDC1 in situ recruitment and activation combined with stemness inhibition provides great immune response and anti-tumor potential, providing new ideas for clinical tumor vaccine design.


Antigen Presentation , Cancer Vaccines , Dendritic Cells , Hydrogels , Cancer Vaccines/immunology , Mice , Animals , Dendritic Cells/immunology , Antigen Presentation/immunology , T-Lymphocytes, Cytotoxic/immunology , Disease Models, Animal , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/drug effects , Humans , Mice, Inbred C57BL
18.
Int Immunopharmacol ; 132: 111972, 2024 May 10.
Article En | MEDLINE | ID: mdl-38569429

The potential of cytotoxic CD4+ T cells and tissue resident memory T cells (Trm) in achieving adult leukemia remission have been highlighted [1,2]. We hypothesized that CXCR6 could serve as a marker for cytotoxic CD4+ Trm cells in the bone marrow (BM) of pediatric B-ALL patients. Flow cytometry (FCM) and published single cell RNA sequencing (scRNA-seq) datasets were employed to characterize CXCR6+CD4+ T cells in the BM and peripheral blood (PB) of pediatric B-ALL patients and healthy donors. FCM, scRNA-seq and co-culture were utilized to explore the cytotoxicity of CXCR6+CD4+ T cells in vitro based on in vitro induction of CXCR6+CD4+ T cells using tumor antigens and peripheral blood mononuclear cells (PBMCs). The ssGSEA based on the cell markers identified according to the in vivo scRNA-seq data, the TARGET-ALL-P2 datasets, and integrated machine learning algorithm were employed to figure out the key cells with prognostic values, followed by simulation of adoptive cell transfer therapy (ACT). Integrated machine learning identified the high-risk cells for disease free survival, and overall survival, while simulation of ACT therapy using CXCR6+CD4+T cells indicated that CXCR6+CD4+ T cells could remodel the bone marrow microenvironments towards anti-tumor. Based on the expression of genes involved in formation of resident memory T cells, CXCR6 is not a marker of resident memory CD4+T cells but defines therapeutic subtypes of CD4+ cytotoxic T cell lineage for pediatric B-ALL.


Immunotherapy, Adoptive , Receptors, CXCR6 , Humans , Immunotherapy, Adoptive/methods , Child , Receptors, CXCR6/genetics , Receptors, CXCR6/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology , T-Lymphocytes, Cytotoxic/immunology , Male , Child, Preschool , Female , CD4-Positive T-Lymphocytes/immunology , Cell Lineage
19.
Anticancer Res ; 44(5): 1877-1883, 2024 May.
Article En | MEDLINE | ID: mdl-38677758

BACKGROUND/AIM: Human gastric cancer stem-like cells (CSCs)/cancer-initiating cells can be identified as aldehyde dehydrogenase-high (ALDHhigh) cells. Cancer immunotherapy employing immune checkpoint blockade has been approved for advanced gastric cancer cases. However, the effectiveness of cancer immunotherapy against gastric CSCs/CICs remains unclear. This study aimed to investigate the susceptibility of gastric CSCs/CICs to immunotherapy. MATERIALS AND METHODS: Gastric CSCs/CICs were isolated as ALDHhigh cells using the human gastric cancer cell line, MKN-45. ALDHhigh clone cells and ALDHlow clone cells were isolated using the ALDEFLUOR assay. ALDH1A1 expression was assessed via qRT-PCR. Sphere-forming ability was evaluated to confirm the presence of CSCs/CICs. A model neoantigen, AP2S1, was over-expressed in ALDHhigh clone cells and ALDHlow clone cells, and susceptibility to AP2S1-specific TCR-T cells was assessed using IFNγ ELISPOT assay. RESULTS: Three ALDHhigh clone cells were isolated from MKN-45 cells. ALDHhigh clone cells exhibited a stable phenotype in in vitro culture for more than 2 months. The High-36 clone cells demonstrated the highest sphere-forming ability, whereas the Low-8 cells showed the lowest sphere-forming ability. High-36 cells exhibited lower expression of HLA-A24 compared to Low-8 cells. TCR-T cells specific for AP2S1 showed lower reactivity to High-36 cells compared to Low-8 cells. CONCLUSION: High-36 cells and Low-8 cells represent novel gastric CSCs/CICs and non-CSCs/CICs, respectively. ALDHhigh CSCs/CICs evade T cells due to lower expression of HLA class 1.


Aldehyde Dehydrogenase 1 Family , Neoplastic Stem Cells , Stomach Neoplasms , T-Lymphocytes, Cytotoxic , Humans , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/pathology , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Aldehyde Dehydrogenase 1 Family/metabolism , Aldehyde Dehydrogenase 1 Family/genetics , Cell Line, Tumor , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Retinal Dehydrogenase/metabolism , Tumor Escape/immunology , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class I/immunology
20.
Front Immunol ; 15: 1376962, 2024.
Article En | MEDLINE | ID: mdl-38562940

Tumors pose a significant global public health challenge, resulting in numerous fatalities annually. CD8+ T cells play a crucial role in combating tumors; however, their effectiveness is compromised by the tumor itself and the tumor microenvironment (TME), resulting in reduced efficacy of immunotherapy. In this dynamic interplay, extracellular vesicles (EVs) have emerged as pivotal mediators, facilitating direct and indirect communication between tumors and CD8+ T cells. In this article, we provide an overview of how tumor-derived EVs directly regulate CD8+ T cell function by carrying bioactive molecules they carry internally and on their surface. Simultaneously, these EVs modulate the TME, indirectly influencing the efficiency of CD8+ T cell responses. Furthermore, EVs derived from CD8+ T cells exhibit a dual role: they promote tumor immune evasion while also enhancing antitumor activity. Finally, we briefly discuss current prevailing approaches that utilize functionalized EVs based on tumor-targeted therapy and tumor immunotherapy. These approaches aim to present novel perspectives for EV-based tumor treatment strategies, demonstrating potential for advancements in the field.


Extracellular Vesicles , Neoplasms , Humans , CD8-Positive T-Lymphocytes , Extracellular Vesicles/metabolism , Neoplasms/metabolism , T-Lymphocytes, Cytotoxic , Tumor Microenvironment
...