Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.101
Filter
Add more filters

Publication year range
1.
J Ethnopharmacol ; 329: 118115, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38580190

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Sijunzi Decoction (SJZD), a traditional Chinese herbal remedy, is frequently employed in the treatment of various cancers, including colon cancer. Previous research suggests that SJZD plays a pivotal role in modulating the immune system and enhancing immunity against tumors. However, the precise role of SJZD in combating colon cancer and its potential molecular functions in regulating natural killer cells remain elusive. AIMS OF THE STUDY: To elucidate the potential mechanism underlying the anticolon cancer effects of SJZD in synergy with natural killer (NK) cells through both in vivo and in vitro experiments. MATERIALS AND METHODS: In vivo experiments: A subcutaneous tumor mouse model of colon cancer and in vivo NK cell depletion experiments were conducted to observe the anticolon cancer effects of SJZD. Flow cytometry assessed immune cell depletion in mouse spleens, while immunohistochemical (IHC) staining detected the expression of apoptotic genes in tumor tissues. In vitro experiments: The mechanism by which SJZD regulates the sensitization of colon cancer cells to NK cells was investigated using real-time polymerase chain reaction (RT-PCR), western blotting (WB), and co-culture experiments with NK cells. RESULTS: Sijunzi Decoction (SJZD) significantly impeded tumor growth in mice; however, NK cell depletion markedly attenuated the tumor-suppressive effect of SJZD. Immunohistochemical (IHC) results indicated that SJZD increased the expression of P53, death receptor 4 (DR4), and death receptor 5 (DR5) in tumor tissues. In vitro experiments, 24 h SJZD-pretreated colon cancer cells showed a substantial elevation in P53, DR4, and DR5 levels, and the activity of colon cancer cells significantly diminished after co-culture with NK cells. These effects of SJZD were reversed with the addition of the P53 inhibitor pifithrin-α (PFT-α), resulting in reduced inhibition of colon cancer cells by NK cells. CONCLUSION: SJZD enhances the levels of DR4 and DR5 through the modulation of P53 expression, consequently increasing the sensitivity of colon cancer cells to NK cell-mediated killing. These findings provide a theoretical foundation for the clinical application of SJZD in patients with colon cancer. In this study, we first investigated the effect of SJZD on subcutaneous tumor growth in mice with colon cancer using in vivo assays and assessed the impact of NK cells on the anticolon cancer effect of SJZD in vivo through NK cell depletion. In vitro experiments were conducted to explore the potential mechanism of action of SJZD in NK cell-mediated anticolon cancer effects.


Subject(s)
Colonic Neoplasms , Drugs, Chinese Herbal , Killer Cells, Natural , Tumor Suppressor Protein p53 , Animals , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Tumor Suppressor Protein p53/metabolism , Drugs, Chinese Herbal/pharmacology , Mice , Humans , Mice, Inbred BALB C , Cell Line, Tumor , Apoptosis/drug effects , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use
2.
Cancer Immunol Res ; 12(6): 744-758, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38526128

ABSTRACT

ω-3 polyunsaturated fatty acids (PUFA) are known to directly repress tumor development and progression. In this study, we explored whether docosahexaenoic acid (DHA), a type of ω-3 PUFA, had an immunomodulatory role in inhibiting tumor growth in immunocompetent mice. The number of natural killer (NK) cells but not the number of T or B cells was decreased by DHA supplementation in various tissues under physiologic conditions. Although the frequency and number of NK cells were comparable, IFNγ production by NK cells in both the spleen and lung was increased in DHA-supplemented mice in the mouse B16F10 melanoma tumor model. Single-cell RNA sequencing revealed that DHA promoted effector function and oxidative phosphorylation in NK cells but had no obvious effects on other immune cells. Using Rag2-/- mice and NK-cell depletion by PK136 antibody injection, we demonstrated that the suppression of B16F10 melanoma tumor growth in the lung by DHA supplementation was dependent mainly on NK cells. In vitro experiments showed that DHA directly enhanced IFNγ production, CD107a expression, and mitochondrial oxidative phosphorylation (OXPHOS) activity and slightly increased proliferator-activated receptor gamma coactivator-1α (PGC-1α) protein expression in NK cells. The PGC-1α inhibitor SR-18292 in vitro and NK cell-specific knockout of PGC-1α in mice reversed the antitumor effects of DHA. In summary, our findings broaden the current knowledge on how DHA supplementation protects against cancer growth from the perspective of immunomodulation by upregulating PGC-1α signaling-mediated mitochondrial OXPHOS activity in NK cells.


Subject(s)
Docosahexaenoic Acids , Killer Cells, Natural , Melanoma, Experimental , Animals , Docosahexaenoic Acids/pharmacology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mice , Melanoma, Experimental/immunology , Melanoma, Experimental/drug therapy , Mice, Knockout , Mice, Inbred C57BL , Interferon-gamma/metabolism , Cell Line, Tumor , Fatty Acids, Omega-3/pharmacology , Oxidative Phosphorylation/drug effects , Humans , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism
3.
Small ; 20(25): e2307521, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38212279

ABSTRACT

Chimeric antigen receptor natural killer (CAR-NK) cell therapy represents a potent approach to suppressing tumor growth because it has simultaneously inherited the specificity of CAR and the intrinsic generality of NK cells in recognizing cancer cells. However, its therapeutic potency against solid tumors is still restricted by insufficient tumor infiltration, immunosuppressive tumor microenvironments, and many other biological barriers. Motivated by the high potency of puerarin, a traditional Chinese medicine extract, in dilating tumor blood vessels, an injectable puerarin depot based on a hydrogen peroxide-responsive hydrogel comprising poly(ethylene glycol) dimethacrylate and ferrous chloride is concisely developed. Upon intratumoral fixation, the as-prepared puerarin depot (abbreviated as puerarin@PEGel) can activate nitrogen oxide production inside endothelial cells and thus dilate tumor blood vessels to relieve tumor hypoxia and reverse tumor immunosuppression. Such treatment can thus promote tumor infiltration, survival, and effector functions of customized epidermal growth factor receptor (HER1)-targeted HER1-CAR-NK cells after intravenous administration. Consequently, such puerarin@PEGel-assisted HER1-CAR-NK cell treatment exhibits superior tumor suppression efficacy toward both HER1-overexpressing MDA-MB-468 and NCI-H23 human tumor xenografts in mice without inducing obvious side effects. This study highlights a potent strategy to activate CAR-NK cells for augmented treatment of targeted solid tumors through reprogramming tumor immunosuppression.


Subject(s)
Immunotherapy , Isoflavones , Killer Cells, Natural , Receptors, Chimeric Antigen , Humans , Animals , Killer Cells, Natural/immunology , Isoflavones/pharmacology , Isoflavones/chemistry , Isoflavones/administration & dosage , Isoflavones/therapeutic use , Immunotherapy/methods , Cell Line, Tumor , Neoplasms/therapy , Mice , Immunosuppression Therapy , Tumor Microenvironment/drug effects , Injections
4.
Proc Natl Acad Sci U S A ; 119(22): e2200568119, 2022 05 31.
Article in English | MEDLINE | ID: mdl-35588144

ABSTRACT

Cyclic dinucleotides (CDN) and Toll-like receptor (TLR) ligands mobilize antitumor responses by natural killer (NK) cells and T cells, potentially serving as complementary therapies to immune checkpoint therapy. In the clinic thus far, however, CDN therapy targeting stimulator of interferon genes (STING) protein has yielded mixed results, perhaps because it initiates responses potently but does not provide signals to sustain activation and proliferation of activated cytotoxic lymphocytes. To improve efficacy, we combined CDN with a half life-extended interleukin-2 (IL-2) superkine, H9-MSA (mouse serum albumin). CDN/H9-MSA therapy induced dramatic long-term remissions of the most difficult to treat major histocompatibility complex class I (MHC I)­deficient and MHC I+ tumor transplant models. H9-MSA combined with CpG oligonucleotide also induced potent responses. Mechanistically, tumor elimination required CD8 T cells and not NK cells in the case of MHC I+ tumors and NK cells but not CD8 T cells in the case of MHC-deficient tumors. Furthermore, combination therapy resulted in more prolonged and more intense NK cell activation, cytotoxicity, and expression of cytotoxic effector molecules in comparison with monotherapy. Remarkably, in a primary autochthonous sarcoma model that is refractory to PD-1 checkpoint therapy, the combination of CDN/H9-MSA with checkpoint therapy yielded long-term remissions in the majority of the animals, mediated by T cells and NK cells. This combination therapy has the potential to activate responses in tumors resistant to current therapies and prevent MHC I loss accompanying acquired resistance of tumors to checkpoint therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Histocompatibility Antigens Class I , Immunotherapy , Interleukin-2 , Membrane Proteins , Neoplasms , Nucleotides, Cyclic , Oligodeoxyribonucleotides , Serum Albumin , Animals , CD8-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class I/genetics , Humans , Immunotherapy/methods , Interleukin-2/immunology , Killer Cells, Natural/immunology , Membrane Proteins/agonists , Mice , Neoplasms/genetics , Neoplasms/therapy , Nucleotides, Cyclic/therapeutic use , Oligodeoxyribonucleotides/therapeutic use , Serum Albumin/therapeutic use
5.
Comput Math Methods Med ; 2022: 9604456, 2022.
Article in English | MEDLINE | ID: mdl-35237344

ABSTRACT

OBJECTIVE: To investigate the potential pharmacological value of extracts from honeysuckle on patients with mild coronavirus disease 2019 (COVID-19) infection. METHODS: The active components and targets of honeysuckle were screened by Traditional Chinese Medicine Database and Analysis Platform (TCMSP). SwissADME and pkCSM databases predict pharmacokinetics of ingredients. The Gene Expression Omnibus (GEO) database collected transcriptome data for mild COVID-19. Data quality control, differentially expressed gene (DEG) identification, enrichment analysis, and correlation analysis were implemented by R toolkit. CIBERSORT evaluated the infiltration of 22 immune cells. RESULTS: The seven active ingredients of honeysuckle had good oral absorption and medicinal properties. Both the active ingredient targets of honeysuckle and differentially expressed genes of mild COVID-19 were significantly enriched in immune signaling pathways. There were five overlapping immunosignature genes, among which RELA and MAP3K7 expressions were statistically significant (P < 0.05). Finally, immune cell infiltration and correlation analysis showed that RELA, MAP3K7, and natural killer (NK) cell are with highly positive correlation and highly negatively correlated with hematopoietic stem cells. CONCLUSION: Our analysis suggested that honeysuckle extract had a safe and effective protective effect against mild COVID-19 by regulating a complex molecular network. The main mechanism was related to the proportion of infiltration between NK cells and hematopoietic stem cells.


Subject(s)
COVID-19 Drug Treatment , Drugs, Chinese Herbal/therapeutic use , Lonicera , Network Pharmacology , Phytotherapy , SARS-CoV-2 , Antiviral Agents/chemistry , Antiviral Agents/pharmacokinetics , Antiviral Agents/therapeutic use , COVID-19/genetics , COVID-19/immunology , Computational Biology , Databases, Pharmaceutical/statistics & numerical data , Drug Evaluation, Preclinical , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/pharmacokinetics , Gene Expression/drug effects , Gene Ontology , Gene Regulatory Networks/drug effects , Gene Regulatory Networks/immunology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/immunology , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Lonicera/chemistry , Medicine, Chinese Traditional , Pandemics , SARS-CoV-2/drug effects
6.
Front Immunol ; 13: 833560, 2022.
Article in English | MEDLINE | ID: mdl-35154155

ABSTRACT

Chagas disease is accompanied by a multisystem inflammatory disorder that follows Trypanosoma cruzi infection. Alpha-tocopherol has been described as an antioxidant and a potential adjuvant to enhance immune responses to vaccines. Therefore, we have evaluated the immune response to T. cruzi infection upon alpha-tocopherol pre-administration. The results show that administration of alpha-tocopherol before the infection results in lower parasitemia and lower mortality of C57BL/6 mice infected with the Tulahuen T. cruzi strain. Alpha-tocopherol administration in normal C57BL/6 mice resulted in higher levels of IFN-γ production by T and NK cells before and after the infection with T. cruzi. More importantly, previous administration of alpha-tocopherol increased the production of IL-10 by T and myeloid suppressor cells and the formation of effector memory T cells while decreasing the expression of PD-1 on T cells. These results suggest that alpha-tocopherol may limit the appearance of dysfunctional T cells during the acute and early chronic phases of T. cruzi infection, contributing to control infection. In addition, alpha-tocopherol could diminish tissue inflammation and fibrosis in late acute disease. These results strongly suggest that alpha-tocopherol may be a helpful agent to be considered in Chagas disease.


Subject(s)
Chagas Disease/prevention & control , Parasitemia/prevention & control , alpha-Tocopherol/pharmacology , Animals , Chagas Disease/pathology , Fibrosis/prevention & control , Inflammation/prevention & control , Interferon-gamma/physiology , Interleukin-10/physiology , Killer Cells, Natural/immunology , Memory T Cells/immunology , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/metabolism
7.
Comput Math Methods Med ; 2022: 8361665, 2022.
Article in English | MEDLINE | ID: mdl-35047059

ABSTRACT

OBJECTIVE: To explore the effect of autoimmune cell therapy on immune cells in patients with chronic obstructive pulmonary disease (COPD) and to provide a reference for clinical treatment of COPD. METHODS: Sixty patients with stable COPD were randomly divided into control group and treatment group (n = 30). The control group was given conventional treatment, and the treatment group was given one autoimmune cell therapy on the basis of conventional treatment. The serum levels of CD3+ T cells, CD4+ T cells, CD8+ cells, B cells, and NK cells in the peripheral blood were detected by flow cytometry. Possible adverse reactions were detected at any time during treatment. RESULTS: There were no significant differences in the contents of CD3+ T cells, CD4+ T cells, CD8+ cells, B cells, and NK cells in the serum of the control group (P > 0.05). Compared with before treatment, the contents of CD3+ T cells, CD4+ T cells, CD8+ cells, B cells, and NK cells in the serum of the treatment group were significantly increased (P < 0.05). The ratio of CD4 + /CD8+ T cells in both control and treatment groups did not change significantly during treatment (P > 0.05). There were no significant differences in serum CD3+ T cells, CD4+ T cells, CD8+ cells, B cells, and NK cells in the treatment group at 30 days and 90 days after treatment (P > 0.05), but they were significantly higher than those in the control group (P < 0.05). CONCLUSION: Autoimmune cell therapy can significantly increase the level of immune cells in the body and can be maintained for a long period of time, which has certain clinical benefits for recurrent respiratory tract infections and acute exacerbation in patients with COPD.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/therapy , Aged , Aged, 80 and over , Autoimmunity , B-Lymphocytes/immunology , B-Lymphocytes/transplantation , Blood Transfusion, Autologous/methods , Blood Transfusion, Autologous/statistics & numerical data , Cell- and Tissue-Based Therapy/statistics & numerical data , Computational Biology , Female , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/transplantation , Leukocyte Transfusion/methods , Leukocyte Transfusion/statistics & numerical data , Male , Middle Aged , Pulmonary Disease, Chronic Obstructive/blood , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/transplantation
8.
Front Immunol ; 12: 765906, 2021.
Article in English | MEDLINE | ID: mdl-34899716

ABSTRACT

Vitamin C (VitC), in addition to its role as a general antioxidant, has long been considered to possess direct anti-cancer activity at high doses. VitC acts through oxidant and epigenetic mechanisms, which at high doses can exert direct killing of tumor cells in vitro and delay tumor growth in vivo. Recently, it has also been shown that pharmacologic-dose VitC can contribute to control of tumors by modulating the immune system, and studies have been done interrogating the role of physiologic-dose VitC on novel adoptive cellular therapies (ACTs). In this review, we discuss the effects of VitC on anti-tumor immune cells, as well as the mechanisms underlying those effects. We address important unanswered questions concerning both VitC and ACTs, and outline challenges and opportunities facing the use of VitC in the clinical setting as an adjunct to immune-based anti-cancer therapies.


Subject(s)
Ascorbic Acid/therapeutic use , Dietary Supplements , Immunotherapy , Neoplasms/therapy , Vitamins/therapeutic use , Animals , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Neoplasms/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
9.
Front Immunol ; 12: 781337, 2021.
Article in English | MEDLINE | ID: mdl-34925361

ABSTRACT

Existing first-line cancer therapies often fail to cope with the heterogeneity and complexity of cancers, so that new therapeutic approaches are urgently needed. Among novel alternative therapies, adoptive cell therapy (ACT) has emerged as a promising cancer treatment in recent years. The limited clinical applications of ACT, despite its advantages over standard-of-care therapies, can be attributed to (i) time-consuming and cost-intensive procedures to screen for potent anti-tumor immune cells and the corresponding targets, (ii) difficulties to translate in-vitro and animal-derived in-vivo efficacies to clinical efficacy in humans, and (iii) the lack of systemic methods for the safety assessment of ACT. Suitable experimental models and testing platforms have the potential to accelerate the development of ACT. Immunocompetent microphysiological systems (iMPS) are microfluidic platforms that enable complex interactions of advanced tissue models with different immune cell types, bridging the gap between in-vitro and in-vivo studies. Here, we present a proof-of-concept iMPS that supports a triple culture of three-dimensional (3D) colorectal tumor microtissues, 3D cardiac microtissues, and human-derived natural killer (NK) cells in the same microfluidic network. Different aspects of tumor-NK cell interactions were characterized using this iMPS including: (i) direct interaction and NK cell-mediated tumor killing, (ii) the development of an inflammatory milieu through enrichment of soluble pro-inflammatory chemokines and cytokines, and (iii) secondary effects on healthy cardiac microtissues. We found a specific NK cell-mediated tumor-killing activity and elevated levels of tumor- and NK cell-derived chemokines and cytokines, indicating crosstalk and development of an inflammatory milieu. While viability and morphological integrity of cardiac microtissues remained mostly unaffected, we were able to detect alterations in their beating behavior, which shows the potential of iMPS for both, efficacy and early safety testing of new candidate ACTs.


Subject(s)
Biological Assay/methods , Cell Culture Techniques, Three Dimensional/methods , Immunotherapy, Adoptive , Killer Cells, Natural/transplantation , Neoplasms/therapy , Biological Assay/instrumentation , Cell Culture Techniques, Three Dimensional/instrumentation , Cell Line , Cell Separation , Female , Fetal Blood , Healthy Volunteers , Humans , Induced Pluripotent Stem Cells , Intravital Microscopy , Killer Cells, Natural/immunology , Lab-On-A-Chip Devices , Male , Myocytes, Cardiac , Neoplasms/immunology , Neoplasms/pathology , Primary Cell Culture , Proof of Concept Study
10.
Front Immunol ; 12: 680611, 2021.
Article in English | MEDLINE | ID: mdl-34956168

ABSTRACT

Natural killer (NK) cells are a potent weapon against tumor and viral infection. Finding active compounds with the capacity of enhancing NK cell effector functions will be effective to develop new anti-cancer drugs. In this study, we initially screened 287 commercially available active compounds by co-culturing with peripheral blood mononuclear cells (PBMCs). We found that five compounds, namely, Daphnetin, MK-8617, LW6, JIB-04, and IOX1, increased the IFN-γ+ NK cell ratio in the presence of IL-12. Further studies using purified human primary NK cells revealed that Daphnetin directly promoted NK cell IFN-γ production in the presence of IL-12 but not IL-15, while the other four compounds acted on NK cells indirectly. Daphnetin also improved the direct cytotoxicity of NK cells against tumor cells in the presence of IL-12. Through RNA-sequencing, we found that PI3K-Akt-mTOR signaling acted as a central pathway in Daphnetin-mediated NK cell activation in the presence of IL-12. This was further confirmed by the finding that both inhibitors of PI3K-Akt and its main downstream signaling mTOR, LY294002, and rapamycin, respectively, can reverse the increase of IFN-γ production and cytotoxicity in NK cells promoted by Daphnetin. Collectively, we identify a natural product, Daphnetin, with the capacity of promoting human NK cell activation via PI3K-Akt-mTOR signaling in the presence of IL-12. Our current study opens up a new potential application for Daphnetin as a complementary immunomodulator for cancer treatments.


Subject(s)
Cytotoxicity, Immunologic/drug effects , Interferon-gamma/biosynthesis , Killer Cells, Natural/drug effects , Lymphocyte Activation/drug effects , Umbelliferones/pharmacology , Acetanilides/pharmacology , Adamantane/analogs & derivatives , Adamantane/pharmacology , Adolescent , Adult , Aminopyridines/pharmacology , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacology , Female , Humans , Hydrazones/pharmacology , Hydroxyquinolines/pharmacology , Interferon-gamma/genetics , Interleukin-12/physiology , K562 Cells , Killer Cells, Natural/immunology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Male , Middle Aged , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , Pyridazines/pharmacology , Pyrimidines/pharmacology , Signal Transduction , TOR Serine-Threonine Kinases/physiology , Young Adult
11.
Molecules ; 26(19)2021 Oct 01.
Article in English | MEDLINE | ID: mdl-34641520

ABSTRACT

Immunotherapy harnessing immune functions is a promising strategy for cancer treatment. Tumor sensitization is one approach to enhance tumor cell susceptibility to immune cell cytotoxicity that can be used in combination with immunotherapy to achieve therapeutic efficiency. Cordycepin, a bioactive compound that can be extracted from some Cordyceps spp. has been reported to effectively inhibit tumor growth, however, the mechanism of its tumor sensitization activity that enhances immune cell cytotoxicity is unknown. In the present study, we investigated the potency of cordycepin to sensitize a lethal cancer, cholangiocarcinoma (CCA), to natural killer (NK) cells. Treatment with cordycepin prior to and during co-culturing with NK-92 cells significantly increased cell death of KKU-213A as compared to solitary cordycepin or NK treatment. Moreover, sensitization activity was also observed in the combination of NK-92 cells and Cordyceps militaris extract that contained cordycepin as a major component. The cordycepin treatment remarkably caused an increase in TRAIL receptor (DR4 and DR5) expression in KKU-213A, suggesting the possible involvement of TRAIL signaling in KKU-213A sensitization to NK-92 cells. In conclusion, this is the first report on the sensitization activity of cordycepin on CCA cells to NK cytotoxicity, which supports that cordycepin can be further developed as an alternate immunomodulating agent.


Subject(s)
Bile Duct Neoplasms/drug therapy , Cholangiocarcinoma/drug therapy , Cordyceps/chemistry , Deoxyadenosines/pharmacology , Killer Cells, Natural/immunology , Antineoplastic Agents, Phytogenic/pharmacology , Bile Duct Neoplasms/pathology , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Cholangiocarcinoma/pathology , Gene Expression Regulation, Neoplastic/drug effects , Histocompatibility Antigens Class I/genetics , Humans , Killer Cells, Natural/drug effects , Plant Extracts/pharmacology , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , fas Receptor/genetics
12.
Int J Mol Sci ; 22(19)2021 Sep 30.
Article in English | MEDLINE | ID: mdl-34638944

ABSTRACT

Natural polysaccharides have shown promising effects on the regulation of immunity in animals. In this study, we examined the immune stimulatory effect of intranasally administered Codium fragile polysaccharides (CFPs) in mice. Intranasal administration of CFPs in C57BL/6 mice induced the upregulation of surface activation marker expression in macrophages and dendritic cells (DCs) in the mediastinal lymph node (mLN) and the production of interleukin-6 (IL-6), IL-12p70, and tumor necrosis factor-α in bronchoalveolar lavage fluid. Moreover, the number of conventional DCs (cDCs) was increased in the mLNs by the upregulation of C-C motif chemokine receptor 7 expression, and subsets of cDCs were also activated following the intranasal administration of CFP. In addition, the intranasal administration of CFPs promoted the activation of natural killer (NK) and T cells in the mLNs, which produce pro-inflammatory cytokines and cytotoxic mediators. Finally, daily administration of CFPs inhibited the infiltration of Lewis lung carcinoma cells into the lungs, and the preventive effect of CFPs on tumor growth required NK and CD8 T cells. Furthermore, CFPs combined with anti-programmed cell death-ligand 1 (PD-L1) antibody (Ab) improved the therapeutic effect of anti-PD-L1 Ab against lung cancer. Therefore, these data demonstrated that the intranasal administration of CFP induced mucosal immunity against lung cancer.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antineoplastic Agents/administration & dosage , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/therapy , Chlorophyta/chemistry , Immunity, Mucosal , Immunotherapy/methods , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Phytotherapy/methods , Plant Extracts/administration & dosage , Polysaccharides/administration & dosage , Administration, Intranasal/methods , Animals , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Lewis Lung/pathology , Cell Line, Tumor , Dendritic Cells/immunology , Disease Models, Animal , Female , Killer Cells, Natural/immunology , Lung Neoplasms/pathology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL
13.
J Immunol Res ; 2021: 9483433, 2021.
Article in English | MEDLINE | ID: mdl-34485538

ABSTRACT

Medicinal plants serve as a lead source of bioactive compounds and have been an integral part of day-to-day life in treating various disease conditions since ancient times. Withaferin A (WFA), a bioactive ingredient of Withania somnifera, has been used for health and medicinal purposes for its adaptogenic, anti-inflammatory, and anticancer properties long before the published literature came into existence. Nearly 25% of pharmaceutical drugs are derived from medicinal plants, classified as dietary supplements. The bioactive compounds in these supplements may serve as chemotherapeutic substances competent to inhibit or reverse the process of carcinogenesis. The role of WFA is appreciated to polarize tumor-suppressive Th1-type immune response inducing natural killer cell activity and may provide an opportunity to manipulate the tumor microenvironment at an early stage to inhibit tumor progression. This article signifies the cumulative information about the role of WFA in modulating antitumor immunity and its potential in targeting prostate cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Prostatic Neoplasms/drug therapy , Withania/chemistry , Withanolides/pharmacology , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Disease Models, Animal , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Male , Prostate/drug effects , Prostate/immunology , Prostate/pathology , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Th1 Cells/drug effects , Th1 Cells/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Withanolides/therapeutic use
14.
Int J Mol Sci ; 22(18)2021 Sep 11.
Article in English | MEDLINE | ID: mdl-34575998

ABSTRACT

During the last decade, we have persistently addressed the question, "how can the innate immune system be used as a therapeutic tool to eliminate cancer?" A cancerous tumor harbors innate immune cells such as macrophages, which are held in the tumor-promoting M2 state by tumor-cell-released cytokines. We have discovered that these tumor-associated macrophages (TAM) are repolarized into the nitric oxide (NO)-generating tumoricidal M1 state by the dietary agent curcumin (CC), which also causes recruitment of activated natural killer (NK) cells and cytotoxic T (Tc) cells into the tumor, thereby eliminating cancer cells as well as cancer stem cells. Indications are that this process may be NO-dependent. Intriguingly, the maximum blood concentration of CC in mice never exceeds nanomolar levels. Thus, our results submit that even low, transient levels of curcumin in vivo are enough to cause repolarization of the TAM and recruitment NK cells as well as Tc cells to eliminate the tumor. We have observed this phenomenon in two cancer models, glioblastoma and cervical cancer. Therefore, this approach may yield a general strategy to fight cancer. Our mechanistic studies have so far implicated induction of STAT-1 in this M2→M1 switch, but further studies are needed to understand the involvement of other factors such as the lipid metabolites resolvins in the CC-evoked anticancer pathways.


Subject(s)
Curcumin/therapeutic use , Glioblastoma/drug therapy , Neoplasms, Experimental/drug therapy , Uterine Cervical Neoplasms/drug therapy , Animals , Female , Glioblastoma/immunology , Glioblastoma/pathology , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Mice , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Nitric Oxide/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/pathology , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/pathology , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/pathology
15.
Nutrients ; 13(9)2021 Aug 24.
Article in English | MEDLINE | ID: mdl-34578808

ABSTRACT

The aim of this study was to re-validate the changes in natural killer (NK) cell cytotoxicity and cytokines related to T cells after Sil-Q1 (SQ; silk peptide) supplementation in a larger pool of Korean adults with minimized daily dose of SQ and controlling seasonal influence compared to the previous study. A total of 130 subjects were randomly assigned (1:1) to consume either 7.5 g of SQ or placebo for 8 weeks. NK cell cytotoxicity and cytokines were measured at T0 (baseline) and T8 (follow-up). Comparing the NK cell cytotoxicity values at T0 and T8 within each group, the cytotoxicity at all effector cell (E) to target cell (T) ratios of 10:1, 5:1, 2.5:1, and 1.25:1 was significantly increased in the SQ group at T8. Additionally, significant differences in the changed value (Δ, subtract baseline values from follow-up values) comparison between the groups at E:T = 10:1, 5:1, and 2.5:1 were found. As a secondary endpoint, the interleukin (IL)-12 level in the SQ group was significantly increased for 8 weeks, and Δ IL-12 in the SQ group was greater than in the placebo group. In conclusion, the present study showed considerable practical implications of SQ supplementation. Thus, SQ is an effective and safe functional food supplement for enhancing immune function.


Subject(s)
Amino Acids/administration & dosage , Cytokines/drug effects , Killer Cells, Natural/drug effects , Peptides/administration & dosage , Silk/administration & dosage , Cytokines/immunology , Dietary Supplements , Female , Functional Food , Humans , Interleukin-12/blood , Killer Cells, Natural/immunology , Korea , Male , Middle Aged , Seasons , Silk/chemistry , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Treatment Outcome
16.
Int J Mol Sci ; 22(17)2021 Aug 24.
Article in English | MEDLINE | ID: mdl-34502035

ABSTRACT

Although fucoidan, a well-studied seaweed-extracted polysaccharide, has shown immune stimulatory effects that elicit anticancer immunity, mucosal adjuvant effects via intranasal administration have not been studied. In this study, the effect of Ecklonia cava-extracted fucoidan (ECF) on the induction of anti-cancer immunity in the lung was examined by intranasal administration. In C57BL/6 and BALB/c mice, intranasal administration of ECF promoted the activation of dendritic cells (DCs), natural killer (NK) cells, and T cells in the mediastinal lymph node (mLN). The ECF-induced NK and T cell activation was mediated by DCs. In addition, intranasal injection with ECF enhanced the anti-PD-L1 antibody-mediated anti-cancer activities against B16 melanoma and CT-26 carcinoma tumor growth in the lungs, which were required cytotoxic T lymphocytes and NK cells. Thus, these data demonstrated that ECF functioned as a mucosal adjuvant that enhanced the immunotherapeutic effect of immune checkpoint inhibitors against metastatic lung cancer.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Immune Checkpoint Inhibitors/therapeutic use , Laminaria/chemistry , Lung Neoplasms/drug therapy , Polysaccharides/therapeutic use , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Administration, Intranasal , Animals , Cell Line, Tumor , Dendritic Cells/drug effects , Dendritic Cells/immunology , Drug Combinations , Female , Immune Checkpoint Inhibitors/administration & dosage , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Lung Neoplasms/pathology , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Metastasis , Plant Extracts , Polysaccharides/administration & dosage , Polysaccharides/pharmacology
17.
Genome Med ; 13(1): 129, 2021 08 11.
Article in English | MEDLINE | ID: mdl-34376232

ABSTRACT

BACKGROUND: Tumor response to therapy is affected by both the cell types and the cell states present in the tumor microenvironment. This is true for many cancer treatments, including immune checkpoint inhibitors (ICIs). While it is well-established that ICIs promote T cell activation, their broader impact on other intratumoral immune cells is unclear; this information is needed to identify new mechanisms of action and improve ICI efficacy. Many preclinical studies have begun using single-cell analysis to delineate therapeutic responses in individual immune cell types within tumors. One major limitation to this approach is that therapeutic mechanisms identified in preclinical models have failed to fully translate to human disease, restraining efforts to improve ICI efficacy in translational research. METHOD: We previously developed a computational transfer learning approach called projectR to identify shared biology between independent high-throughput single-cell RNA-sequencing (scRNA-seq) datasets. In the present study, we test this algorithm's ability to identify conserved and clinically relevant transcriptional changes in complex tumor scRNA-seq data and expand its application to the comparison of scRNA-seq datasets with additional data types such as bulk RNA-seq and mass cytometry. RESULTS: We found a conserved signature of NK cell activation in anti-CTLA-4 responsive mouse and human tumors. In human metastatic melanoma, we found that the NK cell activation signature associates with longer overall survival and is predictive of anti-CTLA-4 (ipilimumab) response. Additional molecular approaches to confirm the computational findings demonstrated that human NK cells express CTLA-4 and bind anti-CTLA-4 antibodies independent of the antibody binding receptor (FcR) and that similar to T cells, CTLA-4 expression by NK cells is modified by cytokine-mediated and target cell-mediated NK cell activation. CONCLUSIONS: These data demonstrate a novel application of our transfer learning approach, which was able to identify cell state transitions conserved in preclinical models and human tumors. This approach can be adapted to explore many questions in cancer therapeutics, enhance translational research, and enable better understanding and treatment of disease.


Subject(s)
CTLA-4 Antigen/antagonists & inhibitors , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Lymphocyte Activation/genetics , Models, Biological , Neoplasms/genetics , Transcriptome , Animals , Biomarkers , Cell Line, Tumor , Computational Biology/methods , Databases, Genetic , Disease Models, Animal , Drug Evaluation, Preclinical , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Ipilimumab/pharmacology , Ipilimumab/therapeutic use , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , Mice , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Prognosis , ROC Curve , Treatment Outcome
18.
J Food Sci ; 86(8): 3492-3504, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34250593

ABSTRACT

Interferon-γ (IFN-γ) regulates the human immune system. To study the interaction between macrophages and natural killer (NK) cells, we established a THP-1 macrophage-conditioned media. Among the 58 natural plant extracts tested, Curcuma longa exerted the strongest IFN-γ-enhancing effect in NK-92 cells through THP-1 macrophages. C. longa extract (CLE) enhanced IFN-γ secretion 2.3- and 4.2-fold at 50 and 100 µg/ml, respectively. Therefore, we evaluated its IFN-γ-enhancing effect in vitro. Although NK-92 cells did not produce IFN-γ following treatment with C. longa, enhanced IFN-γ secretion was observed after treatment with THP-1 macrophage-conditioned media. We hypothesized that the cytokines secreted by the CLE-treated THP-1 macrophages are responsible for stimulating NK-92 cells. Cytokine array results show upregulation of cytokines, including MIP-1α, CXCL-1, IL-1ß, PAI-1, and TNF-α, in CLE-treated THP-1 macrophages. To determine the cytokines responsible for augmenting IFN-γ secretion, NK-92 cells were stimulated with MIP-1α, CXCL-1, IL-1ß, or PAI-1. Enzyme-linked immunosorbent assay results show that all cytokines induced IFN-γ production, although the dose response was somewhat varied. High-performance liquid chromatography analysis of CLE revealed the concentrations of three active curcuminoids, curcumin, demethoxycurcumin, and bisdemethoxycurcumin, as 6.70%, 1.00%, and 0.95%, respectively. Their mixture (with concentrations comparable to their occurrence in CLE) exerted an effect similar to that of the whole CLE. Our findings reveal that CLE indirectly stimulated NK-92 cells to secrete IFN-γ, which is mediated by cytokines produced from THP-1 macrophages. Further, we identified three curcuminoids partly responsible for this IFN-γ-enhancing effect. Therefore, C. longa can be used as a functional food ingredient owing to its immune-boosting ability. PRACTICAL APPLICATION: This study demonstrates that CLE stimulates THP-1 macrophages to secrete cytokines, which can in turn stimulate IFN-γ production by NK-92 cells. A mixture of three curcuminoids present in the extract exerted effects similar to whole CLE, demonstrating that the curcuminoids are partly responsible for the IFN-γ-enhancing effect of C. longa. Since IFN-γ is a key regulator of human immune system, these results suggest the potential use of C. longa as an immune-boosting functional food ingredient.


Subject(s)
Curcuma , Cytokines , Killer Cells, Natural , Macrophages , Plant Extracts , Adjuvants, Immunologic/pharmacology , Curcuma/chemistry , Cytokines/metabolism , Humans , Interferon-gamma/metabolism , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Macrophages/drug effects , Plant Extracts/pharmacology
19.
Clin Exp Immunol ; 206(2): 161-172, 2021 11.
Article in English | MEDLINE | ID: mdl-34331768

ABSTRACT

Gamma-tocotrienol (γT3) is an analogue of vitamin E with beneficial effects on the immune system, including immune-modulatory properties. This study reports the immune-modulatory effects of daily supplementation of γT3 on host T helper (Th) and T regulatory cell (Treg ) populations in a syngeneic mouse model of breast cancer. Female BALB/c mice were fed with either γT3 or vehicle (soy oil) for 2 weeks via oral gavage before they were inoculated with syngeneic 4T1 mouse mammary cancer cells (4T1 cells). Supplementation continued until the mice were euthanized. Mice (n = 6) were euthanized at specified time-points for various analysis (blood leucocyte, cytokine production and immunohistochemistry). Tumour volume was measured once every 7 days. Gene expression studies were carried out on tumour-specific T lymphocytes isolated from splenic cultures. Supplementation with γT3 increased CD4+ (p < 0.05), CD8+ (p < 0.05) T-cells and natural killer cells (p < 0.05) but suppressed Treg cells (p < 0.05) in peripheral blood when compared to animals fed with the vehicle. Higher interferon (IFN)-γ and lower transforming growth factor (TGF)-ꞵ levels were noted in the γT3 fed mice. Immunohistochemistry findings revealed higher infiltration of CD4+ cells, increased expression of interleukin-12 receptor-beta-2 (IL-12ꞵ2R), interleukin (IL)-24 and reduced expression of cells that express the forkhead box P3 (FoxP3) in tumours from the γT3-fed animals. Gene expression studies showed the down-regulation of seven prominent genes in splenic CD4+ T cells isolated from γT3-fed mice. Supplementation with γT3 from palm oil-induced T cell-dependent cell-mediated immune responses and suppressed T cells in the tumour microenvironment in a syngeneic mouse model of breast cancer.


Subject(s)
Dietary Supplements , Lymphocytes, Tumor-Infiltrating/immunology , Mammary Neoplasms, Animal/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment/drug effects , gamma-Tocopherol/pharmacology , Animals , Cell Line, Tumor , Cytokines/immunology , Female , Killer Cells, Natural/immunology , Mammary Neoplasms, Animal/drug therapy , Mice , Mice, Inbred BALB C , Neoplasm Proteins/immunology
20.
Int J Mol Sci ; 22(11)2021 May 31.
Article in English | MEDLINE | ID: mdl-34072732

ABSTRACT

CAR-T (chimeric antigen receptor T) cells have emerged as a milestone in the treatment of patients with refractory B-cell neoplasms. However, despite having unprecedented efficacy against hematological malignancies, the treatment is far from flawless. Its greatest drawbacks arise from a challenging and expensive production process, strict patient eligibility criteria and serious toxicity profile. One possible solution, supported by robust research, is the replacement of T lymphocytes with NK cells for CAR expression. NK cells seem to be an attractive vehicle for CAR expression as they can be derived from multiple sources and safely infused regardless of donor-patient matching, which greatly reduces the cost of the treatment. CAR-NK cells are known to be effective against hematological malignancies, and a growing number of preclinical findings indicate that they have activity against non-hematological neoplasms. Here, we present a thorough overview of the current state of knowledge regarding the use of CAR-NK cells in treating various solid tumors.


Subject(s)
Immunotherapy, Adoptive , Killer Cells, Natural/immunology , Neoplasms/therapy , Receptors, Antigen, T-Cell/immunology , Animals , Antigens, Neoplasm/immunology , Cell Culture Techniques , Clinical Trials as Topic , Combined Modality Therapy/methods , Disease Models, Animal , Drug Evaluation, Preclinical , Genetic Engineering , Humans , Killer Cells, Natural/metabolism , Neoplasms/diagnosis , Neoplasms/etiology , Neoplasms/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL