Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Exp Dermatol ; 32(10): 1823-1833, 2023 10.
Article in English | MEDLINE | ID: mdl-37578092

ABSTRACT

T-LAK cell-oriented protein kinase (TOPK) potently promotes malignant proliferation of tumour cells and is considered as a maker of tumour progression. Psoriasis is a common inflammatory skin disease characterized by abnormal proliferation of keratinocytes. However, the role of TOPK in psoriasis has not been well elucidated. This study aims to investigate the expression and role of TOPK in psoriasis, and the role of TOPK inhibitor in psoriasis attenuation. Gene Expression Omnibus datasets derived from psoriasis patients and psoriatic model mice were screened for analysis. Skin specimens from psoriasis patients were collected for TOPK immunohistochemical staining to investigate the expression and localization of TOPK. Next, psoriatic mice model was established to further confirm TOPK expression pattern. Then, TOPK inhibitor was applied to investigate the role of TOPK in psoriasis progression. Finally, cell proliferation assay, apoptosis assay and cell cycle analysis were performed to investigate the potential mechanism involved. Our study showed that TOPK was upregulated in the lesions of both psoriasis patients and psoriatic model mice, and TOPK levels were positively associated with psoriasis progression. TOPK was upregulated in psoriatic lesions and expressed predominantly by epidermal keratinocytes. In addition, TOPK levels in epidermal keratinocytes were positively correlated with epidermal hyperplasia. Furthermore, topical application of TOPK inhibitor OTS514 obviously alleviated disease severity and epidermal hyperplasia. Mechanismly, inhibiting TOPK induces G2/M phase arrest and apoptosis of keratinocytes, thereby attenuating epidermal hyperplasia and disease progression. Collectively, this study identifies that upregulation of TOPK in keratinocytes promotes psoriatic progression, and inhibiting TOPK attenuates epidermal hyperplasia and psoriatic progression.


Subject(s)
Neoplasms , Psoriasis , Humans , Animals , Mice , Protein Kinase Inhibitors , Hyperplasia/pathology , Killer Cells, Lymphokine-Activated/metabolism , Killer Cells, Lymphokine-Activated/pathology , T-Lymphocytes/metabolism , Keratinocytes/metabolism , Psoriasis/metabolism , Cell Cycle Checkpoints , Apoptosis/genetics , Neoplasms/metabolism , Cell Proliferation/genetics
2.
J Cell Mol Med ; 27(12): 1637-1652, 2023 06.
Article in English | MEDLINE | ID: mdl-37226642

ABSTRACT

KRAS mutation is the most frequent type of genetic mutation in non-small cell lung cancer (NSCLC), especially in lung adenocarcinoma. However, KRAS mutation can affect many biological processes and the mechanisms underlying KRAS mutation-mediate carcinogenesis in NSCLC have not been fully understood. In this research, we found that KRASG12C mutation was associated with the upregulation of T-LAK cell-originated protein kinase (TOPK), which is a well-known serine/threonine MAPK-like protein kinase implicated in tumorigenesis. The overexpression of TOPK significantly promoted the malignant phenotype of A549 cells, and TOPK silencing impaired the malignant phenotype with KRASG12C mutation. Moreover, we demonstrated that TOPK level was regulated by MAPK/ERK signalling and the transcription factor Elk1. TOPK was also found to promote the activation of NF-κB signalling in A549 cells with KRASG12C mutation via facilitating the phosphorylation of TAK1. In the in vivo tumorigenesis model, the administration of TOPK inhibitor OTS514 enhanced the anticancer effect of 5-FU, and the combinatory use of OTS514 and KRASG12C inhibitor AMG510 showed synergistic anti-tumour effect. These results suggest that KRAS-TOPK axis contributes to the progression of NSCLC and targeting this axis could synergize with anticancer effect of the existing chemotherapeutics.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinogenesis/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Killer Cells, Lymphokine-Activated/metabolism , Killer Cells, Lymphokine-Activated/pathology , Lung Neoplasms/pathology , Mutation/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism
3.
Phytother Res ; 37(7): 3057-3068, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36882184

ABSTRACT

Xanthohumol is a principal prenylated chalcone isolated from hops. Previous studies have shown that xanthohumol was effective against various types of cancer, but the mechanisms, especially the direct targets for xanthohumol to exert an anticancer effect, remain elusive. Overexpression of T-lymphokine-activated killer cell-originated protein kinase (TOPK) promotes tumorigenesis, invasion and metastasis, implying the likely potential for targeting TOPK in cancer prevention and treatment. In the present study, we found that xanthohumol significantly inhibited the cell proliferation, migration and invasion of non-small cell lung cancer (NSCLC) in vitro and suppressed tumor growth in vivo, which is well correlated with inactivating TOPK, evidenced by reduced phosphorylation of TOPK and its downstream signaling histone H3 and Akt, and decreased its kinase activity. Moreover, molecular docking and biomolecular interaction analysis showed that xanthohumol was able to directly bind to the TOPK protein, suggesting that TOPK inactivation by xanthohumol is attributed to its ability to directly interact with TOPK. The findings of the present study identified TOPK as a direct target for xanthohumol to exert its anticancer activity, revealing novel insight into the mechanisms underlying the anticancer activity of xanthohumol.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Mitogen-Activated Protein Kinase Kinases/metabolism , Lung Neoplasms/pathology , Molecular Docking Simulation , Killer Cells, Lymphokine-Activated/metabolism , Killer Cells, Lymphokine-Activated/pathology , Cell Line, Tumor
4.
Cell Death Dis ; 13(9): 828, 2022 09 27.
Article in English | MEDLINE | ID: mdl-36167821

ABSTRACT

T-LAK cell-oriented protein kinase (TOPK) is a potential therapeutic target in tumors. However, its role in anaplastic lymphoma kinase (ALK)-positive non-small cell lung cancer (NSCLC) has not been reported. Here, we found that TOPK was highly expressed in ALK-positive NSCLC. Additionally, ALK was identified as another upstream kinase of TOPK by in vitro kinase assay screening. Then, it was proven that ALK phosphorylated TOPK at Y74 in vitro and ex vivo, and the pathways downstream of ALK-TOPK were explored by phosphoproteomic analysis. Subsequently, we demonstrated that inhibiting TOPK enhanced tumor sensitivity to alectinib (an ALK inhibitor). The combination of alectinib and HI-032 (a TOPK inhibitor) suppressed the growth and promoted the apoptosis of ALK-positive NSCLC cells ex vivo and in vivo. Our findings reveal a novel ALK-TOPK signaling pathway in ALK-positive NSCLC. The combination of alectinib and HI-032 might be a promising therapeutic strategy for improving the sensitivity of ALK-positive NSCLC to targeted therapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Anaplastic Lymphoma Kinase/genetics , Apoptosis/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Humans , Killer Cells, Lymphokine-Activated/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein Kinases , Signal Transduction
5.
Exp Cell Res ; 382(1): 111447, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31150612

ABSTRACT

Heterotypic CICs (cell-in-cell structures) have been found between tumor cells and various immune cells in a variety of cancer tissues. The frequency of CICs has been found to correlate with tumor malignancy in some studies but not in others. Herein, we examined in depth the CICs observed in colon cancer to determine their potential significance in disease progression. Heterotypic CICs were observed by histochemistry between epithelial cells and lymphocytes in an expanded spectrum of colon tissue from colitis to cancer and in vitro studies were performed using the colonic tumor cell line HCT8 and human peripheral blood lymphocytes. Our data revealed that the CICs formed by colonic epithelial cells and infiltrated lymphocytes not only positively correlated with tumor malignancy but also were upregulated by the inflammatory cytokine IL-6. In addition, we observed that colon cancer cells could initiate autophagy for survival after cytotoxic lymphocyte internalization and that IL-6 could also be involved in this process to promote the death of lymphocytes in CIC structures. Furthermore, certain changes were observed in tumor cells after experiencing CICs. Our findings suggest that CICs formed by colon cancer cells and lymphocytes contribute to tumor escape from immune surveillance, which could be facilitated by IL-6, and might represent a previously undescribed pathway for tumor cells to adapt and evade host immune defense.


Subject(s)
Autophagy/physiology , Cell-in-Cell Formation/physiology , Colonic Neoplasms/pathology , Interleukin-6/physiology , Tumor Escape/physiology , Adenocarcinoma/pathology , Adenoma/pathology , Autophagosomes/physiology , Cell Line, Tumor , Colitis, Ulcerative/pathology , Disease Progression , Epithelial Cells/pathology , Humans , Killer Cells, Lymphokine-Activated/pathology , Lymphocytes, Tumor-Infiltrating/pathology , Lysosomes/metabolism , Microtubule-Associated Proteins/physiology , Mitochondria/physiology , Reactive Oxygen Species/metabolism , T-Lymphocytes, Cytotoxic/pathology
6.
Curr Mol Med ; 18(5): 335-342, 2018.
Article in English | MEDLINE | ID: mdl-30289072

ABSTRACT

Antibody-toxin fused agents or immunotoxins, are a newly engineered class of cytotoxic agents consisting of a bacterial or plant toxin moiety hooked up either to a monoclonal antibody or a specific growth factor. Nevertheless, acquiring a full potency in clinic is mostly restricted due to the Capillary leak syndrome (CLS), a serious immune provoked, life-threatening side effect, subsequent to the endothelial damage, resulting in fluid escape from the bloodstream into tissues including lungs, muscle and brain, developing organ failure and eventually death. Proposed underlying mechanisms include direct damage to endothelial cells, acute inflammation, Lymphokine-activated killer (LAK) cells engagement, alteration in cell-cell/cell-matrix connectivities and cytoskeletal dysfunction. Very poor biodistribution and heterogeneous extravasation pattern in tumor site result in accumulation of ITs close to the extravasation site, gradual toxin release and initiation of nearby endothelial cells lysis, secretion of pro-inflammatory cytokines, development of acute inflammation and engagement of Lymphokine-activated killer (LAK) cells. Intrinsic immunogenicity of applied toxin moiety is another important determinant of CLS incidence. Toxins with more intrinsic immunogenicity possess more probability for CLS development. Recently, development of new generations of antibodies and mutated toxins with conserved cytotoxicity has partly tapered risk of CLS development. Here, we describe probable mechanisms involved in CLS and introduce some of the recently applied strategies for lessening incidence of CLS as much as possible.


Subject(s)
Capillary Leak Syndrome , Cytokines/immunology , Immunotoxins , Killer Cells, Lymphokine-Activated , Neoplasms , Animals , Capillary Leak Syndrome/chemically induced , Capillary Leak Syndrome/immunology , Capillary Leak Syndrome/pathology , Capillary Leak Syndrome/therapy , Humans , Immunotoxins/adverse effects , Immunotoxins/therapeutic use , Inflammation/chemically induced , Inflammation/immunology , Inflammation/pathology , Inflammation/therapy , Killer Cells, Lymphokine-Activated/immunology , Killer Cells, Lymphokine-Activated/pathology , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/pathology
7.
PLoS One ; 8(3): e59418, 2013.
Article in English | MEDLINE | ID: mdl-23555668

ABSTRACT

Cell-in-cell structures refer to a unique phenomenon that one living cell enters into another living cell intactly, occurring between homotypic tumor cells or tumor (or other tissue cells) and immune cells (named as heterotypic cell-in-cell structure). In the present study, through a large scale of survey we observed that heterotypic cell-in-cell structure formation occurred commonly in vitro with host cells derived from different human carcinomas as well as xenotypic mouse tumor cell lines. Most of the lineages of human immune cells, including T, B, NK cells, monocytes as well as in vitro activated LAK cells, were able to invade tumor cell lines. Poorly differentiated stem cells were capable of internalizing immune cells as well. More significantly, heterotypic tumor/immune cell-in-cell structures were observed in a higher frequency in tumor-derived tissues than those in adjacent tissues. In mouse hepatitis models, heterotypic immune cell/hepatocyte cell-in-cell structures were also formed in a higher frequency than in normal controls. After in vitro culture, different forms of internalized immune cells in heterotypic cell-in-cell structures were observed, with one or multiple immune cells inside host cells undergoing resting, degradation or mitosis. More strikingly, some internalized immune cells penetrated directly into the nucleus of target cells. Multinuclear cells with aneuploid nucleus were formed in target tumor cells after internalizing immune cells as well as in situ tumor regions. Therefore, with the prevalence of heterotypic cell-in-cell structures observed, we suggest that shielding of immune cells inside tumor or inflammatory tissue cells implies the formation of aneuploidy with the increased multinucleation as well as fine-tuning of microenvironment under pathological status, which may define distinct mechanisms to influence the etiology and progress of tumors.


Subject(s)
Aneuploidy , Carcinoma/pathology , Cell Communication/immunology , Cell Transformation, Neoplastic/pathology , Giant Cells/pathology , Neoplasms/pathology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Carcinoma/immunology , Cell Differentiation , Cell Transformation, Neoplastic/immunology , Giant Cells/immunology , Hepatocytes/immunology , Hepatocytes/pathology , Humans , Killer Cells, Lymphokine-Activated/immunology , Killer Cells, Lymphokine-Activated/pathology , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Mice , Mitosis , Neoplasms/immunology , Stem Cells/immunology , Stem Cells/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Tumor Cells, Cultured
8.
Cancer Biother Radiopharm ; 27(7): 442-5, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22731929

ABSTRACT

UNLABELLED: Daily short intravenous interleukin-2 (IL-2) infusions have been developed to decrease toxicity while maintaining the anticancer activity of this agent against melanoma. Such IL-2 schedules have previously been shown to promote lymphokine-activated killer cell (LAK) activity. Famotidine may increase LAK activity by increasing IL-2 internalization by the IL-2 receptor on lymphocytes. Twenty-one patients with metastatic melanoma were treated with IL-2 18 million IU/m² intravenously (i.v.) over 15-30 minutes and famotidine 20 mg i.v. daily for 3 days for 6 consecutive weeks on an outpatient basis. Cycles were repeated every 8 weeks. PATIENT CHARACTERISTICS: 13 males/8 females, median age, 51 (range: 26-79), and median Eastern Cooperative Oncology Group performance status, 1; common metastatic sites: lymph nodes (16), lungs (14), subcutaneous (8), liver (7), and bone (7). Prior systemic therapy: chemotherapy (7); IL-2 (7); and interferon (5). Most common toxicities were myalgia/arthralgia, rigors, nausea/emesis, and mild elevation of liver function tests. No patients required hospitalization for toxicity of therapy. One patient (5%) has had a complete response (ongoing at 29+ months), while 4 other patients (19%) had partial responses (total response rate: 24%; 95% confidence interval: 9%-48%). Responses occurred in lung, spleen, bones, lymph nodes, and subcutaneous sites. Median response duration=20+ months. Outpatient intravenous IL-2 and famotidine has activity in melanoma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Killer Cells, Lymphokine-Activated/metabolism , Melanoma/drug therapy , Adult , Aged , Aged, 80 and over , Famotidine/administration & dosage , Female , Humans , Interleukin-2/administration & dosage , Killer Cells, Lymphokine-Activated/pathology , Male , Melanoma/pathology , Middle Aged , Neoplasm Metastasis , Outpatients , Retrospective Studies
9.
Arthritis Rheum ; 60(6): 1753-63, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19479851

ABSTRACT

OBJECTIVE: To determine the cytotoxicity of natural killer (NK) cells and the level of differentiation of hematopoietic stem cells (HSCs) into NK cells in systemic lupus erythematosus (SLE). METHODS: Patients with SLE (n=108), rheumatoid arthritis (RA; n=90), Behçet's disease (n=39), or ankylosing spondylitis (n=41) and healthy control subjects (n=173) were enrolled in the study. NK cell levels, NK cell cytotoxicities, and lymphokine-activated killer (LAK) activities against K562 cells were measured by flow cytometry. Gene expression was assessed by reverse transcription-polymerase chain reaction. NK cells were differentiated from peripheral blood and bone marrow HSCs in vitro. RESULTS: Percentages and absolute numbers of NK cells, cytotoxicities, and LAK activities were significantly lower in the peripheral blood of SLE and RA patients than in that of healthy controls. In particular, this NK cell deficiency was more prominent in patients with lupus nephritis and those with thrombocytopenia. Notably, purified NK cells derived from SLE patients, but not RA patients, were found to have lower cytotoxicities and LAK activities than those from healthy controls. This defect of NK cells in SLE patients was found to be related to lower numbers of NK precursors and to the down-regulation of perforin and granzyme in NK cells. The proliferative capacity of HSCs, the percentages of NK cells differentiated from HSCs, and NK cell cytotoxicities were significantly lower in SLE patients. CONCLUSION: In SLE patients, circulating levels of NK cells were diminished and their cytotoxicities were impaired. Furthermore, the differentiation of HSCs into NK cells was found to be defective. These abnormalities possibly contribute to immune system dysregulation in SLE.


Subject(s)
Cell Differentiation/physiology , Cytotoxicity, Immunologic/physiology , Killer Cells, Natural/pathology , Killer Cells, Natural/physiology , Lupus Erythematosus, Systemic/pathology , Lupus Erythematosus, Systemic/physiopathology , Adult , Apoptosis/physiology , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/physiopathology , Behcet Syndrome/pathology , Behcet Syndrome/physiopathology , Case-Control Studies , Female , Granzymes/metabolism , Humans , Killer Cells, Lymphokine-Activated/pathology , Killer Cells, Lymphokine-Activated/physiology , Leukocytes, Mononuclear/pathology , Leukocytes, Mononuclear/physiology , Lupus Nephritis/pathology , Lupus Nephritis/physiopathology , Male , Middle Aged , Perforin/metabolism , Receptors, Interleukin-2/metabolism , Spondylitis, Ankylosing/pathology , Spondylitis, Ankylosing/physiopathology , Thrombocytopenia/pathology , Thrombocytopenia/physiopathology
10.
Int Immunol ; 20(7): 841-8, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18469328

ABSTRACT

Donor-derived cytokine-induced killer (CIK) can be infused as adoptive immunotherapy after hematopoietic cell transplant (HCT). Promising results were recently reported in HLA-identical HCT, where mild grafts versus host (GVH) events were observed. To extend this strategy across major HLA barriers (e.g. HLA-haploidentical HCT), further studies on CIK cells' alloreactivity are needed. We hypothesized that alloreactivity and anti-tumor activity of CIK cells segregate within two different cell subsets and could consequently be separated according to CD56 and CD3 expression. We tested CIK cells expanded from seven patients who underwent HCT as treatment of metastatic colorectal cancer. We found that CIK cells maintained their alloreactivity across major HLA barriers when tested as bulk population; after CD56-positive selection, anti-tumor activity was restricted to the CD3+/CD56+ cell fraction and alloreactivity versus HLA-mismatched PBMC was restricted to the CD3+/CD56- cell fraction. Bulk CIK cells from engrafted patients did not exhibit alloreactivity in response to host- or donor-derived PBMC, confirming their low potential for GVH across minor HLA barriers. Moreover, we tested if CIK cells expanded from engrafted patients after HCT were as effective as donor-derived ones and could be considered as an alternative option. The expansion rate and tumor cell killing was comparable to that observed in sibling donors. In conclusion, depletion of CD3+/CD56- cells might reduce the risk of GVH without affecting the tumor-killing capacity and could help extending CIK infusions across major HLA barriers. Engrafted patients after HCT could also be considered as an effective alternative option to donor-derived CIK cells.


Subject(s)
Colorectal Neoplasms/immunology , Cytotoxicity, Immunologic , Histocompatibility Antigens/immunology , Killer Cells, Lymphokine-Activated/immunology , Adult , Aged , Blood Component Transfusion , CD56 Antigen/immunology , Cell Separation , Cells, Cultured , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Female , Graft vs Host Disease/immunology , Graft vs Tumor Effect/immunology , Histocompatibility Antigens/blood , Humans , Killer Cells, Lymphokine-Activated/metabolism , Killer Cells, Lymphokine-Activated/pathology , Lymphocyte Subsets/immunology , Lymphocyte Subsets/transplantation , Male , Middle Aged , Transplantation, Homologous/immunology
11.
Anticancer Res ; 26(5A): 3327-32, 2006.
Article in English | MEDLINE | ID: mdl-17094448

ABSTRACT

In adoptive immunotherapy, in vivo trafficking of adoptively transferred cells, including their accumulation at tumor sites, remains to be further investigated. Tracking of these cells by visualization is useful to clarify antitumor mechanisms and develop new modalities to enhance antitumor capacities. In the present study, an in vivo tracking study was performed using an adoptive transfer model of lymphokine-activated killer (LAK) cells induced from green mice into C57/BL6 mice with B16 melanoma metastases. Green mice are green fluorescent protein (GFP) transgenic mice originating from C57/BL6 mice. All of the tissues, except for erythrocytes and hair, express green fluorescence under excitation light. Although LAK cells in combination with IL-2 potently suppressed pulmonary metastases with survival prolongation, very few LAK cells accumulated in tumor tissues compared to those localized in the spleen, as visualized by fluorescent microscopy and quantitated by flow cytometry. The present method using transfer of green mice-derived cells into parental tumor-bearing mice is simple because there is no need for in vitro labeling and is feasible for the in vivo tracking of effector cells in an adoptive immunotherapy model.


Subject(s)
Green Fluorescent Proteins/pharmacokinetics , Killer Cells, Lymphokine-Activated/pathology , Lung Neoplasms/metabolism , Melanoma, Experimental/pathology , Animals , Flow Cytometry , Interleukin-2/therapeutic use , Killer Cells, Lymphokine-Activated/metabolism , Killer Cells, Lymphokine-Activated/transplantation , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Melanoma, Experimental/drug therapy , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Spleen/metabolism , Spleen/pathology , Survival Rate
12.
J Reprod Immunol ; 68(1-2): 27-37, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16229897

ABSTRACT

T helper (Th)1 cytokine-predominating status and compromised placental vasculature is thought to be central to the pathogenesis of preeclampsia. However, it remains to be clarified how these two phenomena relate to each other. We have reported that lymphokine-activated killer (LAK) cells induced from decidual mononuclear cells (DMCs) with interleukin (IL)-2 expressed in preeclamptic placenta reduced the angiogenic activity of cytotrophoblasts (CTs). The objective of this study was to examine how LAK cells reduced the angiogenic activity of CTs. We investigated the angiogenesis-related molecules released from cultured CTs obtained from first trimester placenta that had been pretreated with either non-activated DMCs or LAK cells from DMCs. The amounts of vascular endothelial growth factor (VEGF), placenta growth factor (PlGF) and their antagonist, soluble fms-like tyrosine-kinase-1 (sFlt-1) released in CT culture media were measured using ELISA. CTs pretreated with LAK cells released more sFlt-1 compared with those pretreated with non-activated lymphocytes, and CTs pretreated with non-activated lymphocytes released more sFlt-1 compared with those without pretreatment. The release of total VEGF and free PlGF from CTs was not altered by pretreatment with DMCs. Thus, in preeclamptic placenta, LAK cells induced from DMCs by co-existing IL-2 may react to the invading CTs and enhance the release of sFlt-1 from CTs without any change of VEGF or PlGF secretion. This might result in the reduction of actual angiogenic potential of the VEGF system in decidua and the placental vascular system might be compromised, which may lead to the development of preeclampsia.


Subject(s)
Decidua/immunology , Killer Cells, Lymphokine-Activated/immunology , Neovascularization, Physiologic/immunology , Pre-Eclampsia/immunology , Trophoblasts/immunology , Vascular Endothelial Growth Factor Receptor-1/immunology , Cells, Cultured , Cytokines/immunology , Decidua/blood supply , Decidua/pathology , Female , Humans , Killer Cells, Lymphokine-Activated/pathology , Pre-Eclampsia/pathology , Pre-Eclampsia/physiopathology , Pregnancy , Trophoblasts/metabolism , Trophoblasts/pathology , Vascular Endothelial Growth Factor Receptor-1/metabolism
13.
Oncol Res ; 13(4): 235-41, 2003.
Article in English | MEDLINE | ID: mdl-12659424

ABSTRACT

Generally, lymphokine-activated killer (LAK) cells kill target cells early after the LAK cells adhere to them. In this study, we describe that LAK cells can also kill at a later time, such as 24-96 h. LAK cells were generated from a cancer patient and healthy volunteers. As target cells, the patient's autotumor cell line H41 was used. When LAK cells were added to the target cells in a culture well, the LAK cells killed the target cells by cell-cell adhesion within 1-4 h (early killing), but not all cells were killed. The LAK cells were then removed. However, the remaining cells ultimately died 24-96 h later (late killing). The late killing was different from the early killing because numerous granules and vacuoles appeared in the cytoplasm. The late killing was not induced by adding supernatant of the LAK cell culture, suggesting that LAK-target cell contact may be necessary for the killing. The cell injury was inhibited by 3-methyladenine (lysosome inhibitor). It suggests that the vacuoles may be caused by activated lysosome. The patient's LAK cells induced late killing at high levels. There was a high percentage of CD8(+)CD16(+) cells in the peripheral blood lymphocytes (PBL). This subset induced late killing more effectively than the CD8(-)CD16(+) subset. Killing was more conspicuous against H41 than against allogeneic cell line T98G. This type of killing is noteworthy for understanding of killing mechanism of LAK cells.


Subject(s)
Cytotoxicity, Immunologic , Killer Cells, Lymphokine-Activated/immunology , Lymphocyte Activation , Lymphocyte Subsets/pathology , Cell Death/drug effects , Cell Death/immunology , Cells, Cultured , Culture Media, Conditioned/pharmacology , Humans , Killer Cells, Lymphokine-Activated/drug effects , Killer Cells, Lymphokine-Activated/pathology , Lymphocyte Subsets/drug effects , Lymphocyte Subsets/immunology , Male , Middle Aged , Tumor Cells, Cultured
14.
Acta Pharmacol Sin ; 23(3): 257-62, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11918852

ABSTRACT

AIM: To investigate the effect of reactive oxygen species on the proliferation of lymphokine-activated killer (LAK) cells in patients with bladder cancer and their cytolysis to bladder tumor cells. METHODS: Sodium nitroprusside (SNP) was used as nitric oxide (NO) donor. The superoxide anion (O2-.) was generated in the complete medium (CM) supplemented with N-methylphenazonium methyl sulfate (PMS) 3-120 micromol/L and nicotinamide adenine dinucleotide (NADH) 18 - 600 micromol/L. The hydroxyl radical (.OH) was produced by adding ascorbic acid (AA) 0.5 - 400 micromol/L and ferrous sulfate (Fe2+) 0.05 - 40 micromol/L in CM. LAK cell proliferation and cytotoxicity were assayed in the presence of NO, .OH, or O2-. Bladder cancer cell lines BIU-87 and EJ were cultured as target cells and cytotoxicity of LAK cells were determined by MTT assay. RESULTS: The proliferation of LAK cells induced by interleukin-2 (IL-2) was inhibited by hydroxyl radical from 48 h to 96 h in a dose-dependent fashion and was inhibited to 34.5 % compared with control at 96 h in the concentration of ascorbic acid 400 micromol/L and ferrous sulfate 40 micromol/L. The inhibition induced by.OH can be overcome by certain concentrations of mannitol or editic acid. On the contrary, the proliferation of LAK cells induced by IL-2 was stimulated by certain concentrations of NO or O2-. The stimulation induced by O2-. can be overcome to control level by superoxide dismutase (SOD) 3 10(5) U/L. Exogenous O2-. resulted in an increase in cytotoxicity of LAK cells against BIU87 and EJ cells. However, the LAK cells cytotoxicity treated with hydroxyl radical or SOD showed no difference as compared with the control. CONCLUSION: NO and O2-. enhanced the proliferation and activation and O2-. up-regulated antitumor cytotoxicity of LAK cells in patients with bladder cancer. The growth of LAK cells induced by IL-2 was down-regulated by hydroxyl radical. The effects of these reactive oxygen species on the proliferation of LAK cells induced by IL-2 were different.


Subject(s)
Carcinoma, Transitional Cell/immunology , Killer Cells, Lymphokine-Activated/drug effects , Reactive Oxygen Species/pharmacology , Urinary Bladder Neoplasms/immunology , Carcinoma, Transitional Cell/pathology , Cell Division/drug effects , Cells, Cultured , Cytotoxicity, Immunologic/drug effects , Humans , Interleukin-2/pharmacology , Killer Cells, Lymphokine-Activated/immunology , Killer Cells, Lymphokine-Activated/pathology , Tumor Cells, Cultured , Urinary Bladder Neoplasms/pathology
15.
Acta Haematol ; 105(4): 209-21, 2001.
Article in English | MEDLINE | ID: mdl-11528095

ABSTRACT

We have already shown that cytokine cocktails (IL-1beta, IL-3, IL-6, SCF, GM-CSF) and/or lymphokine-activated killer (LAK) cells can reduce the amounts of clonal, CD34-positive mononuclear bone marrow cells (BM-MNC) in acute myeloid leukemia (AML). In addition, the influence of those cocktails and/or LAK cells on the clonogenic potential of AML BM-MNC was investigated. BM colonies cultured in agar during different stages of the disease were immunophenotyped in situ: 17 patients at diagnosis, 14 patients in complete remission, 8 patients at relapse, 8 healthy donors. A significant reduction in leukemic cells and colonies positive for CD34 after in vitro culture of BM-MNC with cytokine cocktails was achieved with all samples obtained at diagnosis (n = 8, p < 0.01), in 6 of 8 cases in complete remission but only in 2 of 6 cases at relapse. Cytokine cocktails stimulated granulopoiesis as well as B and T lymphopoiesis. Colonies with leukemic phenotype could never be detected in healthy BM. A significant reduction in leukemic colonies was achieved by coculture of BM-MNC (uncultured or cytokine precultured) with autologous LAK cells in all 4 cases at diagnosis and in 1 case at relapse. An additive effect of in vitro cytokine preincubation of BM samples on the leukemia-reducing effect of LAK cells could be demonstrated in all samples studied (p < 0.001; diagnosis: n = 10, relapse: n = 3, complete remission: n = 7). Patients had a better prognosis if CD34-positive colonies in AML could be reduced by cytokine incubation (p = 0.03) or coculture with autologous LAK cells in vitro (p = 0.04). Our data show that cytokines as well as LAK cells alone and in combination can reduce, however not eliminate clonogenic AML cells. Such mechanisms might be responsible for maintaining stable remissions in AML.


Subject(s)
Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Cytokines/immunology , Killer Cells, Lymphokine-Activated/immunology , Leukemia, Myeloid/immunology , Leukemia, Myeloid/pathology , Acute Disease , Antigens, CD34 , Cell Differentiation , Cell Division , Cytokines/pharmacology , Cytokines/therapeutic use , Cytotoxicity, Immunologic , Female , Humans , Immunotherapy , Killer Cells, Lymphokine-Activated/pathology , Leukemia, Myeloid/mortality , Leukemia, Myeloid/therapy , Male , Middle Aged , Tumor Cells, Cultured
16.
In Vivo ; 14(5): 651-8, 2000.
Article in English | MEDLINE | ID: mdl-11125549

ABSTRACT

The circulatory pattern of IL-2 activated natural killer (A-NK) cells was studied in C57BL/6 mice bearing 10 day-old pulmonary and subcutaneous (s.c.) metastases of the B16 melanoma in order to evaluate the roles of the concentration of A-NK cells in the blood and of tumor blood flow on accumulation of A-NK cells in tumors. Kinetic studies of the presence of A-NK cells in peripheral blood after adoptive transfer revealed that these cells rapidly disappear from the blood. Via intravital microscopy of animals with exposed lung tissue, we have shown that the vast majority of transferred A-NK cells become efficiently arrested within the lung microcirculation at their first encounter with this organ, thereby explaining the fast disappearance of the cells from the bloodstream. Despite the low number of A-NK cells circulating in the blood, systemically injected A-NK cells (20 million per mouse) localized significantly (70-80 million cells/g) into most pulmonary metastases within 8-16 hours. In contrast, very few A-NK cells (< 0.2 million cells/g) were found in the s.c. metastases. Based on measurements of tumor blood flow (showing a classic inverse relationship between tumor size and tumor blood flow) and the blood concentration of A-NK cells, we estimated the highest intratumoral density of A-NK cells that theoretically can be generated by A-NK cells transported to the tumor by way of the blood. In s.c. tumors, the observed density of A-NK cells was at all times lower (10-50 fold) than the estimated density, indicating that only a few percent of the A-NK cells arriving at these tumors become retained in them. In contrast, the observed density of A-NK cells in pulmonary metastases was at all times higher (2-3 fold) than the estimated density. This finding indicates that A-NK cells might not reach the pulmonary metastases solely by way of the blood stream. In conclusion, i.v. injected A-NK cells become immediately entrapped in the lungs and, consequently, circulate poorly. While lung metastases become significantly infiltrated by i.v. injected A-NK cells, metastases in organs down-stream from the lungs become poorly infiltrated. We hypothesize that only a part of the A-NK cells found in lung metastases 8-16 hours following injection reach these metastases by way of the blood-vascular system. They might also migrate into the metastases from the surrounding normal lung tissue.


Subject(s)
Interleukin-2/pharmacology , Killer Cells, Lymphokine-Activated/immunology , Lung Neoplasms/blood supply , Melanoma, Experimental/blood supply , Skin Neoplasms/blood supply , Animals , Cell Count , Immunotherapy, Adoptive , Killer Cells, Lymphokine-Activated/drug effects , Killer Cells, Lymphokine-Activated/pathology , Killer Cells, Lymphokine-Activated/transplantation , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/secondary , Mice , Mice, Inbred C57BL , Microcirculation , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Skin Neoplasms/secondary , Spleen/cytology , Spleen/drug effects
17.
Zhongguo Yao Li Xue Bao ; 20(3): 276-8, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10452107

ABSTRACT

AIM: To study the effects of basic fibroblast growth factor (bFGF) on the proliferation of lymphokine-activated killer (LAK) cells from patients with bladder cancer and LAK cells cytolysis against bladder tumor cells. METHODS: LAK cell proliferation was assayed in the presence of various concentrations of bFGF combined with interleukin-2 (IL-2) by cell count. Cytotoxicity of LAK cells against bladder cancer cell line EJ cells and bladder tumor cells (BTC) from patients was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays. RESULTS: The proliferation of peripheral blood monocytes (PBMC) was inhibited by bFGF 5 micrograms.L-1. bFGF did not affect the stimulation of LAK cells induced by IL-2. The LAK cell numbers in the combination of IL-2 with bFGF were not significantly different compared with that treated with IL-2 alone. bFGF enhanced cytotoxicity of LAK cells against bladder cancer cell line EJ cells or BTC, respectively. CONCLUSION: Although the proliferation of PBMC was inhibited by bFGF, bFGF increased LAK cell cytotoxicity against bladder neoplasm cells.


Subject(s)
Carcinoma, Transitional Cell/immunology , Cytotoxicity, Immunologic/drug effects , Fibroblast Growth Factor 2/pharmacology , Killer Cells, Lymphokine-Activated/immunology , Urinary Bladder Neoplasms/immunology , Carcinoma, Transitional Cell/pathology , Cell Division/drug effects , Humans , Killer Cells, Lymphokine-Activated/pathology , Tumor Cells, Cultured , Urinary Bladder Neoplasms/pathology
18.
J Immunother ; 22(1): 33-40, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9924697

ABSTRACT

We studied the effect of a streptococcal preparation, OK-432, on the cytotoxicity and the proliferation of lymphokine-activated killer (LAK) cells for use in adoptive immunotherapy. Peripheral blood mononuclear cells (PBMC) were derived from healthy donors and patients with malignant brain tumors. We divided PBMC into two groups; these cells then were stimulated with interleukin-2 in the presence or absence of OK-432. OK-432 was added only in the initial 3 days during the 3-week midterm culture period. Then, we compared OK-432-stimulated LAK (OK-LAK) cells with standard LAK (sLAK) cells in terms of their rate of proliferation and cytotoxicity. OK-LAK cells proliferated more rapidly than sLAK cells. The cytotoxicity of OK-LAK cells increased, whereas that of sLAK cells decreased. We also investigated the phenotypic differences between these two types of LAK cells and found that, on day 21, the OK-LAK cells consisted mostly of CD3-CD56+ NK cells, whereas the sLAK cells consisted mostly of CD3+CD56- T cells. The difference in their level of cytotoxic potential might be explained by the difference of predominant phenotype.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/therapy , Cytotoxicity, Immunologic/drug effects , Killer Cells, Lymphokine-Activated/immunology , Picibanil/pharmacology , Antigens, CD/analysis , Brain Neoplasms/drug therapy , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Cell Division/drug effects , Dose-Response Relationship, Drug , Flow Cytometry , Humans , Immunotherapy, Adoptive , Interleukin-2/pharmacology , Killer Cells, Lymphokine-Activated/cytology , Killer Cells, Lymphokine-Activated/drug effects , Killer Cells, Lymphokine-Activated/pathology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/pathology , Lymphocyte Activation , Phenotype , Receptors, Interleukin-2/analysis
19.
Blood ; 93(3): 963-73, 1999 Feb 01.
Article in English | MEDLINE | ID: mdl-9920846

ABSTRACT

Human granzyme H is a neutral serine protease that is expressed predominantly in the lymphokine-activated killer (LAK)/natural killer (NK) compartment of the immune system. The gene that encodes this granzyme is located between the granzyme B and cathepsin G genes on human chromosome 14q11.2. Although the murine orthologue of human granzyme H has not yet been identified, murine granzymes C, D, E, F, and G also lie between the murine granzyme B and cathepsin G genes on murine chromosome 14; murine granzymes C, D, and F are also highly expressed in LAK cells, but minimally in cytotoxic T lymphocytes (CTL). We therefore tested whether the 5' flanking region of human granzyme H contains the cis-acting DNA sequences necessary to target a reporter gene to the LAK/NK compartment of transgenic mice. A 1.2-kb fragment of 5' flanking human granzyme H sequence was linked to an SV40 large T-antigen (TAg) reporter gene and used to create six transgenic founder lines. SV40 TAg was specifically expressed in the LAK cells of these mice, but not in resting T or NK cells, in CTL, or in any other tissues. Most mice eventually developed a fatal illness characterized by massive hepatosplenomegaly and disseminated organ infiltration by large malignant lymphocytes. Cell lines derived from splenic tumors were TAg+ and NK1.1(+) large granular lymphocytes and displayed variable expression of CD3, CD8, and CD16. Although these cell lines contained perforin and expressed granzymes A, B, C, D, and F, they did not exhibit direct cytotoxicity. Collectively, these results suggest that the 5' flanking sequences of the human granzyme H gene target expression to an NK/T progenitor compartment and to activated NK (LAK) cells. Mice and humans may therefore share a regulatory "program" for the transcription of NK/LAK specific granzyme genes.


Subject(s)
Gene Expression Regulation, Enzymologic , Genes , Killer Cells, Lymphokine-Activated/enzymology , Killer Cells, Natural/enzymology , Regulatory Sequences, Nucleic Acid , Serine Endopeptidases/genetics , T-Lymphocytes/enzymology , Animals , Antigens, Polyomavirus Transforming/genetics , Antigens, Polyomavirus Transforming/physiology , Chromosomes, Human, Pair 14/genetics , Enzyme Induction , Female , Genes, Reporter , Granzymes , Humans , Killer Cells, Lymphokine-Activated/pathology , Killer Cells, Natural/pathology , Lymphoproliferative Disorders/genetics , Lymphoproliferative Disorders/immunology , Lymphoproliferative Disorders/pathology , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, SCID , Mice, Transgenic , Neoplastic Stem Cells/metabolism , Recombinant Fusion Proteins/physiology , Serine Endopeptidases/biosynthesis , Spleen/pathology , T-Lymphocytes/pathology
20.
Int J Oncol ; 14(2): 361-6, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9917514

ABSTRACT

The interaction of natural killer (NK) cells with target cells, such as K562, results in NK functional inactivation and apoptosis. The role of NK-activating cytokines, IL-2, IL-12, and IL-15, in the regulation of NK inactivation and programmed cell death by target cells was examined. Purified natural killer cells were obtained from human peripheral blood and either co-incubated with K562 target cells and cytokines or the NK cells were pretreated with cytokines for 18 h prior to co-culture with K562 cells. Sorted NK cells were examined for cytotoxic activity and NK co-cultured with K562 were examined for cytokine secretion, phenotyping and DNA fragmentation. The cytotoxic activity was inhibited and was not alleviated by cytokine treatment. Whereas the cytokine treatment maintained NK cell viability for several days, NK cell viability was decreased significantly in the presence of K562 target cells. Downregulation of CD16 and upregulation of CD69 on NK cells were induced by K562 target cells and no modulation of these antigens was observed with cytokine treatment. A subpopulation of target-treated NK cells succumbed to cell death by apoptosis and cell death was not rescued by the activating cytokines. These findings demonstrate that target-induced functional inactivation and apoptosis of NK cells were not rescued by the activating cytokines IL-2, IL-12, and IL-15 regardless of whether the NK cells were pretreated with cytokines prior to exposure to K562 or the cytokines were added to the NK-K562 mixtures. These results also suggest that signals triggered by the target cells and resulting in NK cell anergy and apoptosis override cytokine-mediated signals for activation, cell proliferation, and survival.


Subject(s)
Apoptosis/immunology , Killer Cells, Lymphokine-Activated/immunology , Killer Cells, Natural/immunology , Leukemia, Erythroblastic, Acute/immunology , Tumor Escape , Cells, Cultured , Coculture Techniques , Cytotoxicity, Immunologic , Humans , Killer Cells, Lymphokine-Activated/pathology , Killer Cells, Natural/pathology , Leukemia, Erythroblastic, Acute/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...