Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35.178
Filtrar
Más filtros

Intervalo de año de publicación
1.
Front Immunol ; 15: 1452810, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39380986

RESUMEN

The formation of a lytic immunological synapse (IS) is crucial for cytotoxic lymphocytes to accurately target and effectively eliminate malignant cells. While significant attention has been focused on the lymphocyte side of the IS, particularly its role as a secretory domain for lytic granules, the cancer cell side of the IS has remained relatively underexplored. Recent findings have revealed that cancer cells can rapidly polarize their actin cytoskeleton toward the IS upon interaction with natural killer (NK) cells, thereby evading NK cell-mediated cytotoxicity. In this Brief Research Report, we present preliminary findings suggesting that actin cytoskeleton remodeling at the cancer cell side of the IS is associated with the targeted secretion of small extracellular vesicles towards the interacting NK cell. We observed that multivesicular bodies (MVBs) preferentially accumulate in the synaptic region in cancer cells exhibiting synaptic accumulation of F-actin, compared to those lacking actin cytoskeleton remodeling. Extracellular immunofluorescence staining revealed increased surface exposure of CD63 at the cancer cell side of the IS, suggestive of the fusion of MVBs with the plasma membrane. This hypothesis was supported by a pH-sensitive probe demonstrating dynamic trafficking of CD63 to the extracellular region of the IS. Collectively, our data support the notion that cancer cells can engage in targeted secretion of extracellular vesicles in response to NK cell attack, underscoring the need for further research into the potential role of this process in facilitating cancer cell immune evasion.


Asunto(s)
Sinapsis Inmunológicas , Células Asesinas Naturales , Humanos , Sinapsis Inmunológicas/metabolismo , Sinapsis Inmunológicas/inmunología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Citoesqueleto de Actina/metabolismo , Neoplasias/inmunología , Neoplasias/patología , Neoplasias/metabolismo , Cuerpos Multivesiculares/metabolismo , Cuerpos Multivesiculares/inmunología , Línea Celular Tumoral , Tetraspanina 30/metabolismo , Actinas/metabolismo , Vesículas Extracelulares/inmunología , Vesículas Extracelulares/metabolismo , Citotoxicidad Inmunológica
2.
Int J Mol Sci ; 25(20)2024 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-39456827

RESUMEN

Only some subpopulations of natural killer (NK) cells have cytotoxic functionality, and the effects of anesthetics on these subpopulations are unknown. This study aimed to evaluate the in vitro effects of various anesthetics, both alone and in combination, on the distribution and cytotoxic function of NK cells and their subpopulations. Peripheral blood mononuclear cells (PBMCs) from eight healthy volunteers were treated for 4 h in vitro with dexmedetomidine, remifentanil, lidocaine, propofol, sevoflurane, and combinations in clinically relevant concentrations or left untreated. Flow cytometry was used to quantify the percentage of sampled NK cells and evaluate their distribution (CD56brightCD16neg, CD56brightCD16dim, CD56dimCD16neg, CD56dimCD16bright, and CD56negCD16bright) and cytotoxicity (Granzyme B (GrzB) and perforin) of NK cell subpopulations. Although the percentage of total NK cells did not change following exposure to anesthesia, the most important cytotoxic subpopulation (CD56dimCD16bright NK cells) decreased after exposure to both propofol (-3.58%, p = 0.045) and sevoflurane (-16.10%, p = 0.008) alone, and most combinations, especially in combination with lidocaine (propofol with lidocaine (-9.66%, p = 0.002) and sevoflurane with lidocaine (-21.90%, p < 0.001)). Dexmedetomidine and remifentanil had no effect on CD56dimCD16bright NK cells. Furthermore, no anesthetic regimen or combination altered the expression of GrzB and perforin in NK cells or NK cell subpopulations. In short, propofol and sevoflurane suppressed the highly cytotoxic phenotype (CD56dimCD16bright) of NK cells, with those exposed to sevoflurane combinations showing greater reductions. Immunosuppression was intensified with the inclusion of lidocaine in the anesthetic regimen.


Asunto(s)
Células Asesinas Naturales , Humanos , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Adulto , Masculino , Sevoflurano/farmacología , Anestésicos/farmacología , Lidocaína/farmacología , Propofol/farmacología , Femenino , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Dexmedetomidina/farmacología , Citotoxicidad Inmunológica/efectos de los fármacos , Antígeno CD56/metabolismo , Citometría de Flujo , Remifentanilo/farmacología
3.
Front Immunol ; 15: 1463736, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39445004

RESUMEN

Purpose: The ability to generate natural killer (NK) cells from induced pluripotent stem cells (iPSCs) has given rise to new possibilities for the large-scale production of homogeneous immunotherapeutic cellular products and opened new avenues towards the creation of "off-the-shelf" cancer immunotherapies. However, the differentiation of NK cells from iPSCs remains poorly understood, particularly regarding the ontogenic landscape of iPSC-derived NK (iNK) cells produced in vitro and the influence that the differentiation strategy employed may have on the iNK profile. Methods: To investigate this question, we conducted a comparative analysis of two sets of iNK cells generated from the same iPSC line using two different protocols: (i) a short-term, clinically compatible feeder-free protocol corresponding to primitive hematopoiesis, and (ii) a lymphoid-based protocol representing the definitive hematopoietic step. Results and discussion: Our work demonstrated that both protocols are capable of producing functional iNK cells. However, the two sets of resulting iNKs exhibited distinct phenotypes and transcriptomic profiles. The lymphoid-based differentiation approach generated iNKs with a more mature and activated profile, which demonstrated higher cytotoxicity against cancer cell lines compared to iNK cells produced under short-term feeder-free conditions suggesting that the differentiation strategy must be considered when designing iNK cell-based adoptive immunotherapies.


Asunto(s)
Diferenciación Celular , Citotoxicidad Inmunológica , Células Madre Pluripotentes Inducidas , Células Asesinas Naturales , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/inmunología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Humanos , Diferenciación Celular/inmunología , Transcriptoma , Línea Celular Tumoral
4.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(9): 769-776, 2024 Sep.
Artículo en Chino | MEDLINE | ID: mdl-39442965

RESUMEN

Objective To investigate that Anti-monosialoganglioside antibody(Anti-GM1) inhibits the natural killing activity of natural killer(NK) cells in nude mice by regulating NK cell differentiation. Methods Multiplex immunofluorescence staining was used to identify the NK cell typing characteristics in human renal cancer tissue. Six male BALB/c mice were selected, and GM1 was injected subcutaneously once a week for three weeks to immunize the mice for the preparation of Anti-GM1. Six BALB/c-Nude nude mice were randomly divided into the control group (CON) and the experimental group (Anti-GM1) using a random number table. The CON group was treated with IgG by continuous intraperitoneal injection for 7 days; the Anti-GM1 group was treated with Anti-GM1 by continuous intraperitoneal injection for 7 days. After 7 days, the nude mice were sacrificed by cervical dislocation, the spleens of the nude mice were taken and made into a single-cell suspension, and the splenocytes were co-cultivated in YAC-1 for 4 hours. Killing viability of the NK cells was detected by LDH assay. Detection was performed using flow cytometry, with NK cell subsets defined based on the expression of CD27 and CD11b, and the expression of CD107a was also assessed. ELISA was used to detect the secretion levels of NK cell interferon-gamma (IFN-γ) and granzyme B (GzmB). Results There was an obvious NK cell infiltration in the human renal cancer tissue specimens, and GzmB was also positively expressed. Anti-GM1 can significantly inhibit the killing activity of NK cells. Compared with the CON group, the Anti-GM1 group can promote the differentiation of NK cells into mature subsets. The level of CD107a in NK cells of the Anti-GM1 group was significantly decreased. The secretion levels of IFN-γ and GzmB in NK cells of the Anti-GM1 group were decreased compared to the CON group. Conclusion NK cells can infiltrate into human renal cancer tissue and can secrete GzmB, exerting natural killing effects. Anti-GM1 can promote the maturation of NK cells but inhibit the killing activity of NK cells, which is related to its inhibition of NK cell CD107a expression and suppression of NK cell IFN-γ and GzmB secretion.


Asunto(s)
Diferenciación Celular , Gangliósido G(M1) , Células Asesinas Naturales , Ratones Endogámicos BALB C , Ratones Desnudos , Animales , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/efectos de los fármacos , Humanos , Masculino , Gangliósido G(M1)/inmunología , Ratones , Neoplasias Renales/inmunología , Citotoxicidad Inmunológica , Anticuerpos/inmunología , Interferón gamma/metabolismo , Interferón gamma/inmunología , Línea Celular Tumoral
5.
Front Immunol ; 15: 1440308, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39439794

RESUMEN

Background: Colorectal cancer (CRC) ranks as the third most prevalent malignant neoplasm in terms of both morbidity and mortality. Within the tumor microenvironment (TME) of CRC, the diminished presence and diminished cytotoxic function of natural killer (NK) cells serve as important factors driving the advancement of CRC; however, the precise regulatory mechanisms governing this phenomenon remain incompletely understood. Consequently, the identification of novel, potential anti-CRC targets associated with NK cells emerges as a pressing and paramount concern warranting immediate attention. Methods: We examined the regulatory mechanism of SMAD4-mediated NK cell cytotoxicity on CRC by utilizing various experimental techniques, such as qRT-PCR, flow cytometry. Results: Our findings revealed that the expression of SMAD4 is decreased in NK cells within the TME of human CRC. Furthermore, we observed that enforced upregulation of SMAD4 resulted in enhanced cytotoxicity of NK cells towards CRC cells. Furthermore, our research has revealed that YTHDF2 functions as a downstream effector of SMAD4, playing a crucial role in the control of transcription and translation of m6A-modified RNA. Moreover, our investigation demonstrated that increased expression of SMAD4 promoted the activating receptor NKG2D by elevating levels of YTHDF2. Ultimately, the SMAD4-YTHDF2 regulatory axis significantly enhanced the cytotoxicity of NK cells against human CRC cells. Conclusion: Our study unveils a novel mechanism through which SMAD4 modulates the cytotoxicity of NK cells towards CRC cells, suggesting that SMAD4 may hold promise as a potential therapeutic target for NK cell therapy in CRC.


Asunto(s)
Neoplasias Colorrectales , Citotoxicidad Inmunológica , Células Asesinas Naturales , Proteínas de Unión al ARN , Proteína Smad4 , Microambiente Tumoral , Humanos , Proteína Smad4/genética , Proteína Smad4/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Microambiente Tumoral/inmunología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Adenosina/metabolismo
6.
Front Immunol ; 15: 1470130, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39450161

RESUMEN

Introduction: Adoptive cell therapy using TCR-engineered T-cells is one of the most effective strategies against tumor cells. The TCR T-cell approach has been well tested against a variety of blood neoplasms but is yet to be deeply tested against solid tumors. Among solid tumors, cancer-testis antigens are the most prominent targets for tumor-specific therapy, as they are usually found on cells that lie behind blood-tissue barriers. Methods: We have employed a novel efficient protocol for MAGE-A3-specific T-cell clonal expansion, performed single-cell multi-omic analysis of the expanded T-cells via BD Rhapsody, engineered a selected T-cell receptor into a lentiviral construct, and tested it in an in vitro LDH-cytotoxicity test. Results and discussion: We have observed a 191-fold increase in the MAGE-A3-specific T-cell abundance, obtained a dominant T-cell receptor via single-cell multi-omic BD Rhapsody data analysis in the TCRscape bioinformatics tool, and observed potent cytotoxicity of the dominant-clonotype transduced TCR T-cells against a MAGE-A3-positive tumor. We have demonstrated the efficiency of our T-cell enrichment protocol in obtaining potent anti-tumor T-cells and their T-cell receptors, especially when paired with the modern single-cell analysis methods.


Asunto(s)
Antígenos de Neoplasias , Células Dendríticas , Inmunoterapia Adoptiva , Proteínas de Neoplasias , Receptores de Antígenos de Linfocitos T , Análisis de la Célula Individual , Antígenos de Neoplasias/inmunología , Humanos , Proteínas de Neoplasias/inmunología , Proteínas de Neoplasias/genética , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Inmunoterapia Adoptiva/métodos , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Análisis de la Célula Individual/métodos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Línea Celular Tumoral , Células Clonales , Proliferación Celular , Neoplasias/inmunología , Neoplasias/terapia , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/metabolismo , Citotoxicidad Inmunológica , Multiómica
7.
Cancer Immunol Immunother ; 73(12): 239, 2024 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-39358647

RESUMEN

Natural killer (NK) cells are frontline defenders against cancer and are capable of recognizing and eliminating tumor cells without prior sensitization or antigen presentation. Due to their unique HLA mismatch tolerance, they are ideal for adoptive cell therapy (ACT) because of their ability to minimize graft-versus-host-disease risk. The therapeutic efficacy of NK cells is limited in part by inhibitory immune checkpoint receptors, which are upregulated upon interaction with cancer cells and the tumor microenvironment. Overexpression of inhibitory receptors reduces NK cell-mediated cytotoxicity by impairing the ability of NK cells to secrete effector cytokines and cytotoxic granules. T-cell immunoreceptor with immunoglobulin and ITIM domains (TIGIT), a well-known checkpoint receptor involved in T-cell exhaustion, has recently been implicated in the exhaustion of NK cells. Overcoming TIGIT-mediated inhibition of NK cells may allow for a more potent antitumor response following ACT. Here, we describe a novel approach to TIGIT inhibition using self-delivering RNAi compounds (INTASYL™) that incorporates the features of RNAi and antisense technology. INTASYL compounds demonstrate potent activity and stability, are rapidly and efficiently taken up by cells, and can be easily incorporated into cell product manufacturing. INTASYL PH-804, which targets TIGIT, suppresses TIGIT mRNA and protein expression in NK cells, resulting in increased cytotoxic capacity and enhanced tumor cell killing in vitro. Delivering PH-804 to NK cells before ACT has emerged as a promising strategy to counter TIGIT inhibition, thereby improving the antitumor response. This approach offers the potential for more potent off-the-shelf products for adoptive cell therapy, particularly for hematological malignancies.


Asunto(s)
Inmunoterapia Adoptiva , Células Asesinas Naturales , Neoplasias , Receptores Inmunológicos , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/trasplante , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Receptores Inmunológicos/inmunología , Inmunoterapia Adoptiva/métodos , Neoplasias/terapia , Neoplasias/inmunología , Citotoxicidad Inmunológica , Interferencia de ARN , Línea Celular Tumoral , ARN Interferente Pequeño/genética
8.
J Transl Med ; 22(1): 929, 2024 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-39396988

RESUMEN

BACKGROUND: Chimeric antigen receptor (CAR)-NK cell therapy has shown remarkable clinical efficacy and safety in the treatment of hematological malignancies. However, this efficacy was limited in solid tumors owing to hostile tumor microenvironment (TME). Radiotherapy is commonly used for solid tumors and proved to improve the TME. Therefore, the combination with radiotherapy would be a potential strategy to improve therapeutic efficacy of CAR-NK cells for solid tumors. METHODS: Glypican-3 (GPC3) was used as a target antigen of CAR-NK cell for hepatocellular carcinoma (HCC). To promote migration towards HCC, CXCR2-armed CAR-NK92 cells targeting GPC3 were first developed, and their cytotoxic and migration activities towards HCC cells were evaluated. Next, the effects of irradiation on the anti-tumor activity of CAR-NK92 cells were assessed in vitro and in HCC-bearing NCG mice. Lastly, to demonstrate the potential mechanism mediating the sensitized effect of irradiation on CAR-NK cells, the differential gene expression profiles induced by irradiation were analyzed and the expression of some important ligands for the NK-cell activating receptors were further determined by qRT-PCR and flow cytometry. RESULTS: In this study, we developed CXCR2-armed GPC3-targeting CAR-NK92 cells that exhibited specific and potent killing activity against HCC cells and the enhanced migration towards HCC cells. Pretreating HCC cells with irradiation enhanced in vitro anti-HCC effect and migration activity of CXCR2-armed CAR-NK92 cells. We further found that only high-dose (8 Gy) but not low-dose (2 Gy) irradiation in one fraction could significantly enhanced in vivo anti-HCC activity of CXCR2-armed CAR-NK92 cells. Irradiation with 8 Gy significantly up-regulated the expression of NK cell-activating ligands on HCC cells. CONCLUSIONS: Our results indicate the evidence that irradiation could efficiently enhance the anti-tumor effect of CAR-NK cells in solid tumor model. The combination with radiotherapy would be an attractive strategy to improve therapeutic efficacy of CAR-NK cells for solid tumors.


Asunto(s)
Carcinoma Hepatocelular , Movimiento Celular , Células Asesinas Naturales , Neoplasias Hepáticas , Receptores Quiméricos de Antígenos , Carcinoma Hepatocelular/radioterapia , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/terapia , Carcinoma Hepatocelular/inmunología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/efectos de la radiación , Neoplasias Hepáticas/radioterapia , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/terapia , Neoplasias Hepáticas/inmunología , Animales , Humanos , Línea Celular Tumoral , Receptores Quiméricos de Antígenos/metabolismo , Movimiento Celular/efectos de la radiación , Glipicanos/metabolismo , Receptores de Interleucina-8B/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Ratones , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Inmunoterapia Adoptiva/métodos , Microambiente Tumoral/efectos de la radiación , Citotoxicidad Inmunológica/efectos de la radiación
9.
Sci Rep ; 14(1): 24199, 2024 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-39406778

RESUMEN

Natural Killer (NK) cells are pivotal in immune responses to viral infections, malignancies, autoimmune diseases, and transplantation. Assessment of NK cell adhesion, migration, and cytotoxicity is fundamental for in vitro studies. We propose a novel live-cell tracking method that addresses these three major aspects of NK cell function using human NK cells and primary porcine aortic endothelial cells (PAECs) in two-dimensional (2D) static assays and an in-house cylindrical 3D microfluidic system. The results showed a significant increase of NK cytotoxicity against pTNF-activated PAECs, with apoptotic cell death observed in the majority of dead cells, while no difference was observed in the conventional Delfia assay. Computed analysis of NK cell trajectories revealed distinct migratory behaviors, including trajectory length, diameter, average speed, and arrest coefficient. In 3D microfluidic experiments, NK cell attachment to pTNF-activated PAECs substantially increased, accompanied by more dead PAECs compared to control conditions. NK cell trajectories showed versatile migration in various directions and interactions with PAECs. This study uniquely demonstrates NK attachment and killing in a 3D system that mimics blood vessel conditions. Our microscope method offers sensitive single-cell level results, addressing diverse aspects of NK functions. It is adaptable for studying other immune and target cells, providing insights into various biological questions.


Asunto(s)
Movimiento Celular , Células Endoteliales , Células Asesinas Naturales , Animales , Células Asesinas Naturales/inmunología , Porcinos , Humanos , Adhesión Celular , Citotoxicidad Inmunológica , Microfluídica/métodos , Microfluídica/instrumentación , Células Cultivadas , Comunicación Celular/inmunología , Apoptosis , Rastreo Celular/métodos
10.
Front Immunol ; 15: 1427519, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39328404

RESUMEN

Background: The relationship between chronic hepatitis B (CHB) infection and natural killer (NK) cell dysfunction is well-established, but the specific role of HBV viral antigens in driving NK cell impairment in patients with CHB remains unclear. This study investigates the modulatory effects of hepatitis B virus subviral particles (HBVsvp, a representative model for HBsAg) on the phenotypic regulation (activating and inhibitory receptors), cytokine production and cytotoxic potential of peripheral blood mononuclear cell-derived natural killer cells (PBMCs-derived NK cell), which contributes to NK cell dysfunction in CHB infection, potentially serving as an effective HBV immune evasion strategy by the virus. Methods: NK cells were isolated from peripheral blood of patients with CHB (n=5) and healthy individuals (n=5), stimulated with HBVsvp. Subsequent flow cytometric characterization involved assessing changes in activating (NKp46 and NKG2D) and inhibitory (CD94) receptors expression, quantifying TNF-α and IFN- γ cytokine secretion, and evaluating the cytotoxic response against HepG2.2.15 cells with subsequent HBVsvp quantification. Results: In CHB patients, in vitro exposure of PBMCs-derived NK cell with HBVsvp (represent HBsAg model) significantly reduced NK cell-activating receptors expression (P = 0.022), increased expression of CD94 + NK cells (p = 0.029), accompanied with a reduced TNF-α - IFN-γ cytokine levels, and impaired cytotoxic capacity (evidenced by increased cell proliferation and elevated HBVsvp levels in co-cultures with HepG2.2.15 cells in a time-dependent), relative to healthy donors. Conclusion: These findings suggest that HBVsvp may induce dysfunctional NK cell responses characterized by phenotypic imbalance with subsequent reduction in cytokine and cytotoxic levels, indicating HBVsvp immunosuppressive effect that compromises antiviral defense in CHB patients. These data enhance our understanding of NK cell interactions with HBsAg and highlight the potential for targeting CD94 inhibitory receptors to restore NK cell function as an immunotherapeutic approach. Further clinical research is needed to validate these observations and establish their utility as reliable biomarkers.


Asunto(s)
Virus de la Hepatitis B , Hepatitis B Crónica , Células Asesinas Naturales , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Hepatitis B Crónica/inmunología , Hepatitis B Crónica/virología , Virus de la Hepatitis B/inmunología , Adulto , Masculino , Femenino , Vigilancia Inmunológica , Fenotipo , Persona de Mediana Edad , Citocinas/metabolismo , Citocinas/inmunología , Células Hep G2 , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Citotoxicidad Inmunológica , Antígenos de Superficie de la Hepatitis B/inmunología , Antígenos de Superficie de la Hepatitis B/sangre , Subfamília D de Receptores Similares a Lectina de las Células NK/inmunología , Subfamília D de Receptores Similares a Lectina de las Células NK/metabolismo , Receptor 1 Gatillante de la Citotoxidad Natural/metabolismo
11.
J Innate Immun ; 16(1): 451-469, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39265537

RESUMEN

INTRODUCTION: The serine/threonine with-no-lysine (WNK) kinase family function in blood pressure control, electrolyte homeostasis, and cellular osmoregulation. These kinases and their downstream effectors are considered promising therapeutic targets in hypertension and stroke. However, the role of WNK kinases in immune cells remains poorly understood. METHODS: Using the small-molecule WNK kinase inhibitors WNK463 and WNK-IN-11, we investigated how WNK kinase inhibition affects natural killer (NK) cell physiology. RESULTS: WNK kinase inhibition with WNK463 or WNK-IN-11 significantly decreased IL-2-activated NK cell volume, motility, and cytolytic activity. Treatment of NK cells with these inhibitors induced autophagy by activating AMPK and inhibiting mTOR signaling. Moreover, WNK kinase inhibition increased phosphorylation of Akt and c-Myc by misaligning activity of activating kinases and inhibitory phosphatases. Treatment of tumor-bearing mice with WNK463 impaired tumor metastasis control by adoptively transferred NK cells. CONCLUSION: The catalytic activity of WNK kinases has a critical role of multiple aspects of NK cell physiology and their pharmacologic inhibition negatively impacts NK cell function.


Asunto(s)
Células Asesinas Naturales , Osmorregulación , Células Asesinas Naturales/inmunología , Animales , Ratones , Humanos , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteína Quinasa Deficiente en Lisina WNK 1/metabolismo , Metástasis de la Neoplasia , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Ratones Endogámicos C57BL , Línea Celular Tumoral , Citotoxicidad Inmunológica
12.
Cancer Res Commun ; 4(10): 2673-2684, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39292167

RESUMEN

The molecular mechanisms regulating CD8+ cytotoxic T lymphocytes (CTL) are not fully understood. Here, we show that the peroxisome proliferator-activated receptor δ (PPARδ) suppresses CTL cytotoxicity by inhibiting RelA DNA binding. Treatment of ApcMin/+ mice with the PPARδ agonist GW501516 reduced the activation of normal and tumor-associated intestinal CD8+ T cells and increased intestinal adenoma burden. PPARδ knockout or knockdown in CTLs increased their cytotoxicity against colorectal cancer cells, whereas overexpression of PPARδ or agonist treatment decreased it. Correspondingly, perforin, granzyme B, and IFNγ protein and mRNA levels were higher in PPARδ knockout or knockdown CTLs and lower in PPARδ overexpressing or agonist-treated CTLs. Mechanistically, we found that PPARδ binds to RelA, interfering with RelA-p50 heterodimer formation in the nucleus, thereby inhibiting its DNA binding in CTLs. Thus, PPARδ is a critical regulator of CTL effector function. Significance: Here, we provide the first direct evidence that PPARδ plays a critical role in suppressing the immune response against tumors by downregulating RelA DNA-binding activity. This results in decreased expression of perforin, granzyme B, and IFNγ. Thus, PPARδ may serve as a valuable target for developing future cancer immunotherapies.


Asunto(s)
Linfocitos T CD8-positivos , Granzimas , Perforina , Factor de Transcripción ReIA , Animales , Factor de Transcripción ReIA/metabolismo , Ratones , Granzimas/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Perforina/metabolismo , Perforina/genética , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Linfocitos T Citotóxicos/efectos de los fármacos , PPAR delta/metabolismo , PPAR delta/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Humanos , Citotoxicidad Inmunológica , ADN/metabolismo , Interferón gamma/metabolismo , Línea Celular Tumoral , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Proteínas Citotóxicas Formadoras de Poros/genética
13.
Nat Cancer ; 5(10): 1494-1514, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39261676

RESUMEN

Despite recent advances in immunotherapies targeting single tumor-associated antigens, patients with multiple myeloma eventually relapse. ISB 2001 is a CD3+ T cell engager (TCE) co-targeting BCMA and CD38 designed to improve cytotoxicity against multiple myeloma. Targeting of two tumor-associated antigens by a single TCE resulted in superior cytotoxic potency across a variable range of BCMA and CD38 tumor expression profiles mimicking natural tumor heterogeneity, improved resistance to competing soluble factors and exhibited superior cytotoxic potency on patient-derived samples and in mouse models. Despite the broad expression of CD38 across human tissues, ISB 2001 demonstrated a reduced T cell activation profile in the absence of tumor cells when compared to TCEs targeting CD38 only. To determine an optimal first-in-human dose for the ongoing clinical trial ( NCT05862012 ), we developed an innovative quantitative systems pharmacology model leveraging preclinical data, using a minimum pharmacologically active dose approach, therefore reducing patient exposure to subefficacious doses of therapies.


Asunto(s)
ADP-Ribosil Ciclasa 1 , Mieloma Múltiple , Escape del Tumor , Animales , Humanos , Ratones , ADP-Ribosil Ciclasa 1/inmunología , Antígenos de Neoplasias/inmunología , Antígeno de Maduración de Linfocitos B/inmunología , Línea Celular Tumoral , Citotoxicidad Inmunológica/efectos de los fármacos , Inmunoterapia/métodos , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Mieloma Múltiple/inmunología , Mieloma Múltiple/tratamiento farmacológico , Linfocitos T/inmunología , Linfocitos T/efectos de los fármacos , Escape del Tumor/efectos de los fármacos , Escape del Tumor/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto , Ensayos Clínicos como Asunto
14.
Cancer Immunol Immunother ; 73(11): 221, 2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-39235531

RESUMEN

BACKGROUND: Neuroblastoma is the most common extracranial solid tumor in children and accounts for 15% of pediatric cancer related deaths. Targeting neuroblastoma with immunotherapies has proven challenging due to a paucity of immune cells in the tumor microenvironment and the release of immunosuppressive cytokines by neuroblastoma tumor cells. We hypothesized that combining an oncolytic Herpes Simplex Virus (oHSV) with natural killer (NK) cells might overcome these barriers and incite tumor cell death. METHODS: We utilized MYCN amplified and non-amplified neuroblastoma cell lines, the IL-12 expressing oHSV, M002, and the human NK cell line, NK-92 MI. We assessed the cytotoxicity of NK cells against neuroblastoma with and without M002 infection, the effects of M002 on NK cell priming, and the impact of M002 and priming on the migratory capacity and CD107a expression of NK cells. To test clinical applicability, we then investigated the effects of M002 and NK cells on neuroblastoma in vivo. RESULTS: NK cells were more attracted to neuroblastoma cells that were infected with M002. There was an increase in neuroblastoma cell death with the combination treatment of M002 and NK cells both in vitro and in vivo. Priming the NK cells enhanced their cytotoxicity, migratory capacity and CD107a expression. CONCLUSIONS: To the best of our knowledge, these investigations are the first to demonstrate the effects of an oncolytic virus combined with self-maintaining NK cells in neuroblastoma and the priming effect of neuroblastoma on NK cells. The current studies provide a deeper understanding of the relation between NK cells and neuroblastoma and these data suggest that oHSV increases NK cell cytotoxicity towards neuroblastoma.


Asunto(s)
Células Asesinas Naturales , Neuroblastoma , Viroterapia Oncolítica , Neuroblastoma/terapia , Neuroblastoma/inmunología , Células Asesinas Naturales/inmunología , Humanos , Viroterapia Oncolítica/métodos , Animales , Ratones , Línea Celular Tumoral , Virus Oncolíticos/inmunología , Citotoxicidad Inmunológica , Simplexvirus/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Nat Cell Biol ; 26(10): 1734-1744, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39223375

RESUMEN

Diverse cellular insults converge on activation of the heat shock factor 1 (HSF1), which regulates the proteotoxic stress response to maintain protein homoeostasis. HSF1 regulates numerous gene programmes beyond the proteotoxic stress response in a cell-type- and context-specific manner to promote malignancy. However, the role(s) of HSF1 in immune populations of the tumour microenvironment remain elusive. Here, we leverage an in vivo model of HSF1 activation and single-cell transcriptomic tumour profiling to show that augmented HSF1 activity in natural killer (NK) cells impairs cytotoxicity, cytokine production and subsequent anti-tumour immunity. Mechanistically, HSF1 directly binds and regulates the expression of key mediators of NK cell effector function. This work demonstrates that HSF1 regulates the immune response under the stress conditions of the tumour microenvironment. These findings have important implications for enhancing the efficacy of adoptive NK cell therapies and for designing combinatorial strategies including modulators of NK cell-mediated tumour killing.


Asunto(s)
Factores de Transcripción del Choque Térmico , Células Asesinas Naturales , Microambiente Tumoral , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Factores de Transcripción del Choque Térmico/metabolismo , Factores de Transcripción del Choque Térmico/genética , Animales , Microambiente Tumoral/inmunología , Humanos , Ratones , Ratones Endogámicos C57BL , Línea Celular Tumoral , Citotoxicidad Inmunológica , Citocinas/metabolismo , Regulación Neoplásica de la Expresión Génica , Femenino , Neoplasias/inmunología , Neoplasias/patología , Neoplasias/metabolismo , Neoplasias/genética
16.
J Immunol ; 213(9): 1318-1328, 2024 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-39291926

RESUMEN

The human-derived NK-92 cell-based CAR-NK therapy exhibits inconsistency with overall suboptimal efficacy and rapid in vivo clearance of CAR-NK92 cells in cancer patients. Analysis indicates that although pre-existing IgM in healthy individuals (n = 10) strongly recognizes both NK-92 and CAR-NK92 cells, IgG and IgE do not. However, only a subset of cancer patients (3/8) exhibit strong IgM recognition of these cells, with some (2/8) showing pre-existing IgG recognition. These results suggest a natural immunoreactivity between NK-92 and CAR-NK92 cells and pre-existing human Abs. Furthermore, the therapy's immunogenicity is evidenced by enhanced IgG and IgM recognition postinfusion of CAR-NK92 cells. We also confirmed that healthy plasma's cytotoxicity toward these cells is reduced by complement inhibitors, suggesting that Abs may facilitate the rapid clearance of CAR-NK92 cells through complement-dependent cytotoxicity. Given that NK-92 cells lack known receptors for IgG and IgM, identifying and modifying the recognition targets for these Abs on NK-92 and CAR-NK92 cells may improve clinical outcomes. Moreover, we discovered that the 72nd amino acid of the NKG2D receptor on NK-92 cells is alanine. Previous studies have demonstrated polymorphism at the 72nd amino acid of the NKG2D on human NK cells, with NKG2D72Thr exhibiting a superior activation effect on NK cells compared with NKG2D72Ala. We confirmed this conclusion also applies to NK-92 cells by in vitro cytotoxicity experiments. Therefore, reducing the immunoreactivity and immunogenicity of CAR-NK92 and directly switching NK-92 bearing NKG2D72Ala to NKG2D72Thr represent pressing challenges in realizing NK-92 cells as qualified universal off-the-shelf cellular therapeutics.


Asunto(s)
Inmunoglobulina G , Células Asesinas Naturales , Humanos , Células Asesinas Naturales/inmunología , Inmunoglobulina G/inmunología , Inmunoglobulina M/inmunología , Inmunoterapia Adoptiva/métodos , Citotoxicidad Inmunológica , Neoplasias/inmunología , Neoplasias/terapia , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/genética , Línea Celular Tumoral
17.
Front Biosci (Landmark Ed) ; 29(8): 293, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-39206895

RESUMEN

The tumor microenvironment plays a critical role in modulating immune responses associated with tumorigenesis, tumor progression, and metastasis. Dendritic cells (DC) play a key role in preventing and progression of metastatic neoplasia by driving and restoring dysfunctional immune systems and obliterating immunosuppression, thus obstructing tumor evasion. In this review, we will discuss the functions of tumor-infiltrating DC in anti-tumor resistance, prevention of tumor recurrence, and immunosuppression. We will also describe DC metabolism, differentiation, and plasticity, which are essential for its function. Cancers like Lymphomas may be able to corrupt immune surveillance by reducing natural killer cell numbers. Thus, interactions between lymphoma and DC with reference to cytotoxicity may be an important event, likely to be mediated via activation with interferon-γ (IFN-γ) and Toll like receptors (TLR) ligands. Mechanisms of DC-mediated cytotoxicity and the role of apoptosis and death receptors, including the role played by nitric oxide, etc., are of immense significance. We will also look into the molecular mechanisms in the tumor microenvironment, reduced drug sensitivity, and tumor relapse, as well as methods for combating drug resistance and focusing on immunosuppressive tumor networks. We will address how DC mediated cytotoxicity in combination with drugs affects tumor growth and expansion in relation to checkpoint inhibitors and regulatory T cells. Innovative approaches for therapeutic modulation of this immunosuppressive adoptive DC immunotherapy will be highlighted, which is necessary for future personalized therapeutic applications.


Asunto(s)
Células Dendríticas , Neoplasias , Microambiente Tumoral , Células Dendríticas/inmunología , Humanos , Microambiente Tumoral/inmunología , Microambiente Tumoral/efectos de los fármacos , Neoplasias/inmunología , Neoplasias/terapia , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Animales , Citotoxicidad Inmunológica
18.
Nat Immunol ; 25(10): 1830-1844, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39198632

RESUMEN

The efficacy of antitumor immunity is associated with the metabolic state of cytotoxic T cells, which is sensitive to the tumor microenvironment. Whether ionic signals affect adaptive antitumor immune responses is unclear. In the present study, we show that there is an enrichment of sodium in solid tumors from patients with breast cancer. Sodium chloride (NaCl) enhances the activation state and effector functions of human CD8+ T cells, which is associated with enhanced metabolic fitness. These NaCl-induced effects translate into increased tumor cell killing in vitro and in vivo. Mechanistically, NaCl-induced changes in CD8+ T cells are linked to sodium-induced upregulation of Na+/K+-ATPase activity, followed by membrane hyperpolarization, which magnifies the electromotive force for T cell receptor (TCR)-induced calcium influx and downstream TCR signaling. We therefore propose that NaCl is a positive regulator of acute antitumor immunity that might be modulated for ex vivo conditioning of therapeutic T cells, such as CAR T cells.


Asunto(s)
Citotoxicidad Inmunológica , Receptores de Antígenos de Linfocitos T , Cloruro de Sodio , Microambiente Tumoral , Microambiente Tumoral/inmunología , Humanos , Animales , Ratones , Femenino , Cloruro de Sodio/farmacología , Receptores de Antígenos de Linfocitos T/metabolismo , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Activación de Linfocitos/inmunología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Transducción de Señal
19.
Adv Exp Med Biol ; 1448: 129-144, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39117812

RESUMEN

Familial forms of hemophagocytic lymphohistiocytosis (HLH) are caused by loss-of-function mutations in genes encoding perforin as well as those required for release of perforin-containing cytotoxic granule constituent. Perforin is expressed by subsets of CD8+ T cells and NK cells, representing lymphocytes that share mechanism of target cell killing yet display distinct modes of target cell recognition. Here, we highlight recent findings concerning the genetics of familial HLH that implicate CD8+ T cells in the pathogenesis of HLH and discuss mechanistic insights from animal models as well as patients that reveal how CD8+ T cells may contribute to or drive disease, at least in part through release of IFN-γ. Intriguingly, CD8+ T cells and NK cells may act differentially in severe hyperinflammatory diseases such as HLH. We also discuss how CD8+ T cells may promote or drive pathology in other cytokine release syndromes (CSS). Moreover, we review the molecular mechanisms underpinning CD8+ T cell-mediated lymphocyte cytotoxicity, key to the development of familial HLH. Together, recent insights to the pathophysiology of CSS in general and HLH in particular are providing promising new therapeutic targets.


Asunto(s)
Linfocitos T CD8-positivos , Síndrome de Liberación de Citoquinas , Linfohistiocitosis Hemofagocítica , Humanos , Linfocitos T CD8-positivos/inmunología , Linfohistiocitosis Hemofagocítica/inmunología , Linfohistiocitosis Hemofagocítica/genética , Animales , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/genética , Células Asesinas Naturales/inmunología , Perforina/genética , Perforina/metabolismo , Citotoxicidad Inmunológica/genética , Interferón gamma/inmunología , Interferón gamma/genética , Interferón gamma/metabolismo
20.
J Transl Med ; 22(1): 737, 2024 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-39103915

RESUMEN

BACKGROUND: Cancer stem-like cells (CSCs) play an important role in initiation and progression of aggressive cancers, including esophageal cancer. Natural killer (NK) cells are key effector lymphocytes of innate immunity that directly attack a wide variety of cancer cells. NK cell-based therapy may provide a new treatment option for targeting CSCs. In this study, we aimed to investigate the sensitivity of human esophageal CSCs to NK cell-mediated cytotoxicity. METHODS: CSCs were enriched from human esophageal squamous cell carcinoma cell lines via sphere formation culture. Human NK cells were selectively expanded from the peripheral blood of healthy donors. qRT-PCR, flow cytometry and ELISA assays were performed to examine RNA expression and protein levels, respectively. CFSE-labeled target cells were co-cultured with human activated NK cells to detect the cytotoxicity of NK cells by flow cytometry. RESULTS: We observed that esophageal CSCs were more resistant to NK cell-mediated cytotoxicity compared with adherent counterparts. Consistently, esophageal CSCs showed down-regulated expression of ULBP-1, a ligand for NK cells stimulatory receptor NKG2D. Knockdown of ULBP-1 resulted in significant inhibition of NK cell cytotoxicity against esophageal CSCs, whereas ULBP-1 overexpression led to the opposite effect. Finally, the pro-differentiation agent all-trans retinoic acid was found to enhance the sensitivity of esophageal CSCs to NK cell cytotoxicity. CONCLUSIONS: This study reveals that esophageal CSCs are more resistant to NK cells through down-regulation of ULBP-1 and provides a promising approach to promote the activity of NK cells targeting esophageal CSCs.


Asunto(s)
Citotoxicidad Inmunológica , Regulación hacia Abajo , Neoplasias Esofágicas , Células Asesinas Naturales , Células Madre Neoplásicas , Humanos , Células Asesinas Naturales/inmunología , Neoplasias Esofágicas/patología , Neoplasias Esofágicas/inmunología , Neoplasias Esofágicas/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Regulación hacia Abajo/efectos de los fármacos , Línea Celular Tumoral , Citotoxicidad Inmunológica/efectos de los fármacos , Proteínas Ligadas a GPI/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA