1
|
Lajiness JD, Bloodworth JC, Blankenship RL, Kosins AE, Cook-Mills JM. Dendritic cell-specific deletion of PKCδ in offspring of allergic mothers prevents the predisposition for development of allergic lung inflammation in offspring. J Leukoc Biol 2024; 116:1432-1445. [PMID: 39312649 PMCID: PMC11599121 DOI: 10.1093/jleuko/qiae207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 09/22/2024] [Indexed: 09/25/2024] Open
Abstract
In humans and in mice, maternal allergy predisposes offspring to development of allergy. In murine models, increased levels of maternal β-glucosylceramides are both necessary and sufficient for the development of allergic predisposition in offspring. Furthermore, increased numbers of CD11b+ dendritic cell subsets in the offspring of allergic mothers are associated with allergic predisposition. In vitro, β-glucosylceramides increase CD11b+ dendritic cell subset numbers through increased PKCδ signaling, but it is not known if enhanced PKCδ signaling in dendritic cells is required in vivo. We demonstrate that dendritic cell-specific deletion of PKCδ prevents the β-glucosylceramide-induced increase in CD11b+ dendritic cell subset numbers both in vitro as well as in vivo in the fetal liver of offspring of mothers injected with β-glucosylceramides. Furthermore, dendritic cell-specific deletion of PKCδ in offspring prevents the maternal allergy-induced increase in CD11b+ dendritic cell subsets and decreases allergen-induced interleukin-5 and eosinophilia in lungs of offspring. However, loss of PKCδ in dendritic cells did not prevent development of allergen-specific IgE. Our study provides mechanistic insight into the function of PKCδ in the origins of allergic disease beginning in utero as well as in the development of postnatal allergic lung inflammation.
Collapse
Affiliation(s)
- Jacquelyn D Lajiness
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, United States
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Jeffrey C Bloodworth
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Ross L Blankenship
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Allison E Kosins
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Joan M Cook-Mills
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, United States
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| |
Collapse
|
2
|
Waheed‐Ullah Q, Wilsdon A, Abbad A, Rochette S, Bu'Lock F, Hitz M, Dombrowsky G, Cuello F, Brook JD, Loughna S. Effect of deletion of the protein kinase PRKD1 on development of the mouse embryonic heart. J Anat 2024; 245:70-83. [PMID: 38419169 PMCID: PMC11161829 DOI: 10.1111/joa.14033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 03/02/2024] Open
Abstract
Congenital heart disease (CHD) is the most common congenital anomaly, with an overall incidence of approximately 1% in the United Kingdom. Exome sequencing in large CHD cohorts has been performed to provide insights into the genetic aetiology of CHD. This includes a study of 1891 probands by our group in collaboration with others, which identified three novel genes-CDK13, PRKD1, and CHD4, in patients with syndromic CHD. PRKD1 encodes a serine/threonine protein kinase, which is important in a variety of fundamental cellular functions. Individuals with a heterozygous mutation in PRKD1 may have facial dysmorphism, ectodermal dysplasia and may have CHDs such as pulmonary stenosis, atrioventricular septal defects, coarctation of the aorta and bicuspid aortic valve. To obtain a greater appreciation for the role that this essential protein kinase plays in cardiogenesis and CHD, we have analysed a Prkd1 transgenic mouse model (Prkd1em1) carrying deletion of exon 2, causing loss of function. High-resolution episcopic microscopy affords detailed morphological 3D analysis of the developing heart and provides evidence for an essential role of Prkd1 in both normal cardiac development and CHD. We show that homozygous deletion of Prkd1 is associated with complex forms of CHD such as atrioventricular septal defects, and bicuspid aortic and pulmonary valves, and is lethal. Even in heterozygotes, cardiac differences occur. However, given that 97% of Prkd1 heterozygous mice display normal heart development, it is likely that one normal allele is sufficient, with the defects seen most likely to represent sporadic events. Moreover, mRNA and protein expression levels were investigated by RT-qPCR and western immunoblotting, respectively. A significant reduction in Prkd1 mRNA levels was seen in homozygotes, but not heterozygotes, compared to WT littermates. While a trend towards lower PRKD1 protein expression was seen in the heterozygotes, the difference was only significant in the homozygotes. There was no compensation by the related Prkd2 and Prkd3 at transcript level, as evidenced by RT-qPCR. Overall, we demonstrate a vital role of Prkd1 in heart development and the aetiology of CHD.
Collapse
Affiliation(s)
- Qazi Waheed‐Ullah
- School of Life Sciences, Faculty of Medicine and Health SciencesUniversity of NottinghamNottinghamUK
| | - Anna Wilsdon
- School of Life Sciences, Faculty of Medicine and Health SciencesUniversity of NottinghamNottinghamUK
| | - Aseel Abbad
- School of Life Sciences, Faculty of Medicine and Health SciencesUniversity of NottinghamNottinghamUK
| | - Sophie Rochette
- School of Life Sciences, Faculty of Medicine and Health SciencesUniversity of NottinghamNottinghamUK
| | - Frances Bu'Lock
- East Midlands Congenital Heart CentreUniversity Hospitals of Leicester NHS TrustLeicesterUK
| | - Marc‐Phillip Hitz
- Institute of Medical GeneticsCarl von Ossietzky University OldenburgOldenburgGermany
| | - Gregor Dombrowsky
- Institute of Medical GeneticsCarl von Ossietzky University OldenburgOldenburgGermany
| | - Friederike Cuello
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research CenterUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/LübeckUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - J. David Brook
- School of Life Sciences, Faculty of Medicine and Health SciencesUniversity of NottinghamNottinghamUK
| | - Siobhan Loughna
- School of Life Sciences, Faculty of Medicine and Health SciencesUniversity of NottinghamNottinghamUK
| |
Collapse
|
3
|
Kazanietz MG, Cooke M. Protein kinase C signaling "in" and "to" the nucleus: Master kinases in transcriptional regulation. J Biol Chem 2024; 300:105692. [PMID: 38301892 PMCID: PMC10907189 DOI: 10.1016/j.jbc.2024.105692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 02/03/2024] Open
Abstract
PKC is a multifunctional family of Ser-Thr kinases widely implicated in the regulation of fundamental cellular functions, including proliferation, polarity, motility, and differentiation. Notwithstanding their primary cytoplasmic localization and stringent activation by cell surface receptors, PKC isozymes impel prominent nuclear signaling ultimately impacting gene expression. While transcriptional regulation may be wielded by nuclear PKCs, it most often relies on cytoplasmic phosphorylation events that result in nuclear shuttling of PKC downstream effectors, including transcription factors. As expected from the unique coupling of PKC isozymes to signaling effector pathways, glaring disparities in gene activation/repression are observed upon targeting individual PKC family members. Notably, specific PKCs control the expression and activation of transcription factors implicated in cell cycle/mitogenesis, epithelial-to-mesenchymal transition and immune function. Additionally, PKCs isozymes tightly regulate transcription factors involved in stepwise differentiation of pluripotent stem cells toward specific epithelial, mesenchymal, and hematopoietic cell lineages. Aberrant PKC expression and/or activation in pathological conditions, such as in cancer, leads to profound alterations in gene expression, leading to an extensive rewiring of transcriptional networks associated with mitogenesis, invasiveness, stemness, and tumor microenvironment dysregulation. In this review, we outline the current understanding of PKC signaling "in" and "to" the nucleus, with significant focus on established paradigms of PKC-mediated transcriptional control. Dissecting these complexities would allow the identification of relevant molecular targets implicated in a wide spectrum of diseases.
Collapse
Affiliation(s)
- Marcelo G Kazanietz
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| | - Mariana Cooke
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
4
|
Galbiati V, Lefevre MA, Maddalon A, Vocanson M, Iulini M, Marinovich M, Corsini E. Role of miR-24-3p and miR-146a-5p in dendritic cells' maturation process induced by contact sensitizers. Arch Toxicol 2023; 97:2183-2191. [PMID: 37326882 PMCID: PMC10322961 DOI: 10.1007/s00204-023-03542-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/06/2023] [Indexed: 06/17/2023]
Abstract
MiRNAs are non-coding RNA molecules that regulate gene expression at the post-transcriptional level. Although allergic contact dermatitis has been studied extensively, few studies addressed miRNA expression and their role in dendritic cell activation. The main aim of this work was to investigate the role of miRNAs in the underlying mechanism of dendritic cell maturation induced by contact sensitizers of different potency. Experiments were conducted using THP-1-derived immature DCs (iDCs). Contact allergens of different potency were used: p-benzoquinone, Bandrowski's base, and 2,4-dinitrochlorobenzene as extreme; nickel sulfate hexahydrate, diethyl maleate and 2-mercaptobenzothiazole as moderate; and α-hexyl cinnamaldehyde, eugenol, and imidazolidinyl urea as weak. Selective inhibitor and mimic miRNAs were then used and several cell surface markers was evaluated as targets. Also, patients patch tested with nickel were analyzed to determine miRNAs expression. Results indicate an important role of miR-24-3p and miR-146a-5p in DCs activation. miR-24-3p was up-regulated by extreme and weak contact allergens, while miR-146a-5p was up-regulated by weak and moderate contact allergens and down-regulated only by the extreme ones. Also, the involvement of PKCβ in contact allergen-induced miR-24-3p and miR-146a-5p expression was demonstrated. Furthermore, the expression of the two miRNAs maintains the same trend of expression in both in vitro and in human conditions after nickel exposure. Results obtained suggest the involvement of miR-24 and miR-146a in DCs maturation process in the proposed in vitro model, supported also by human evidences.
Collapse
Affiliation(s)
- Valentina Galbiati
- Laboratory of Toxicology, DiSFeB, Università degli Studi di Milano, Milan, Italy.
| | - Marine-Alexia Lefevre
- CIRI, Centre International de Recherche en Infectiologie, (Team Epidermal Immunity and Allergy), Univ Lyon; Inserm, U1111, Université Claude Bernard Lyon 1; CNRS, UMR5308; ENS de Lyon, Lyon, France
| | - Ambra Maddalon
- Laboratory of Toxicology, DiSFeB, Università degli Studi di Milano, Milan, Italy
| | - Marc Vocanson
- CIRI, Centre International de Recherche en Infectiologie, (Team Epidermal Immunity and Allergy), Univ Lyon; Inserm, U1111, Université Claude Bernard Lyon 1; CNRS, UMR5308; ENS de Lyon, Lyon, France
| | - Martina Iulini
- Laboratory of Toxicology, DiSFeB, Università degli Studi di Milano, Milan, Italy
| | - Marina Marinovich
- Laboratory of Toxicology, DiSFeB, Università degli Studi di Milano, Milan, Italy
| | - Emanuela Corsini
- Laboratory of Toxicology, DiSFeB, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
5
|
Oliver L, Alvarez R, Diaz R, Valdés A, Colligan SH, Nemeth MJ, Twum DYF, Fernández A, Fernández-Medina O, Carlson LM, Yu H, Eng KH, Hensen ML, Rábade-Chediak ML, Fernández LE, Lee KP, Perez L, Muhitch JB, Mesa C, Abrams SI. Mitigating the prevalence and function of myeloid-derived suppressor cells by redirecting myeloid differentiation using a novel immune modulator. J Immunother Cancer 2022; 10:e004710. [PMID: 36150744 PMCID: PMC9511656 DOI: 10.1136/jitc-2022-004710] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/16/2022] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Immune suppression is common in neoplasia and a major driver is tumor-induced myeloid dysfunction. Yet, overcoming such myeloid cell defects remains an untapped strategy to reverse suppression and improve host defense. Exposure of bone marrow progenitors to heightened levels of myeloid growth factors in cancer or following certain systemic treatments promote abnormal myelopoiesis characterized by the production of myeloid-derived suppressor cells (MDSCs) and a deficiency in antigen-presenting cell function. We previously showed that a novel immune modulator, termed 'very small size particle' (VSSP), attenuates MDSC function in tumor-bearing mice, which was accompanied by an increase in dendritic cells (DCs) suggesting that VSSP exhibits myeloid differentiating properties. Therefore, here, we addressed two unresolved aspects of the mechanism of action of this unique immunomodulatory agent: (1) does VSSP alter myelopoiesis in the bone marrow to redirect MDSC differentiation toward a monocyte/macrophage or DC fate? and (2) does VSSP mitigate the frequency and suppressive function of human tumor-induced MDSCs? METHODS To address the first question, we first used a murine model of granulocyte-colony stimulating factor-driven emergency myelopoiesis following chemotherapy-induced myeloablation, which skews myeloid output toward MDSCs, especially the polymorphonuclear (PMN)-MDSC subset. Following VSSP treatment, progenitors and their myeloid progeny were analyzed by immunophenotyping and MDSC function was evaluated by suppression assays. To strengthen rigor, we validated our findings in tumor-bearing mouse models. To address the second question, we conducted a clinical trial in patients with metastatic renal cell carcinoma, wherein 15 patients were treated with VSSP. Endpoints in this study included safety and impact on PMN-MDSC frequency and function. RESULTS We demonstrated that VSSP diminished PMN-MDSCs by shunting granulocyte-monocyte progenitor differentiation toward monocytes/macrophages and DCs with heightened expression of the myeloid-dependent transcription factors interferon regulatory factor-8 and PU.1. This skewing was at the expense of expansion of granulocytic progenitors and rendered the remaining MDSCs less suppressive. Importantly, these effects were also demonstrated in a clinical setting wherein VSSP monotherapy significantly reduced circulating PMN-MDSCs, and their suppressive function. CONCLUSIONS Altogether, these data revealed VSSP as a novel regulator of myeloid biology that mitigates MDSCs in cancer patients and reinstates a more normal myeloid phenotype that potentially favors immune activation over immune suppression.
Collapse
Affiliation(s)
- Liliana Oliver
- Department of Immunoregulation, Immunology and Immunotherapy Direction, Center of Molecular Immunology, Havana, Cuba
| | - Rydell Alvarez
- Department of Immunoregulation, Immunology and Immunotherapy Direction, Center of Molecular Immunology, Havana, Cuba
| | - Raquel Diaz
- Department of Oncology, Joaquín Albarrán Hospital, Havana, Cuba
| | - Anet Valdés
- Department of Immunoregulation, Immunology and Immunotherapy Direction, Center of Molecular Immunology, Havana, Cuba
| | - Sean H Colligan
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Michael J Nemeth
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Danielle Y F Twum
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Audry Fernández
- Department of Immunoregulation, Immunology and Immunotherapy Direction, Center of Molecular Immunology, Havana, Cuba
| | - Olivia Fernández-Medina
- Department of Immunoregulation, Immunology and Immunotherapy Direction, Center of Molecular Immunology, Havana, Cuba
| | - Louise M Carlson
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
- Department of Medicine, Indiana University Simon Comprehensive Cancer Center, Indianapolis, Indiana, USA
| | - Han Yu
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Kevin H Eng
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Mary L Hensen
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Maura L Rábade-Chediak
- Department of Immunoregulation, Immunology and Immunotherapy Direction, Center of Molecular Immunology, Havana, Cuba
| | - Luis Enrique Fernández
- Department of Immunoregulation, Immunology and Immunotherapy Direction, Center of Molecular Immunology, Havana, Cuba
| | - Kelvin P Lee
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
- Department of Medicine, Indiana University Simon Comprehensive Cancer Center, Indianapolis, Indiana, USA
| | - Leslie Perez
- Clinical Direction, Center of Molecular Immunology, Havana, Cuba
| | - Jason B Muhitch
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Circe Mesa
- Department of Immunoregulation, Immunology and Immunotherapy Direction, Center of Molecular Immunology, Havana, Cuba
- Innovative Immunotherapy Alliance, S. A. Mariel, Artemisa, Cuba
| | - Scott I Abrams
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| |
Collapse
|
6
|
Cook-Mills JM, Averill SH, Lajiness JD. Asthma, allergy and vitamin E: Current and future perspectives. Free Radic Biol Med 2022; 179:388-402. [PMID: 34785320 PMCID: PMC9109636 DOI: 10.1016/j.freeradbiomed.2021.10.037] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 10/12/2021] [Accepted: 10/21/2021] [Indexed: 02/03/2023]
Abstract
Asthma and allergic disease result from interactions of environmental exposures and genetics. Vitamin E is one environmental factor that can modify development of allergy early in life and modify responses to allergen after allergen sensitization. Seemingly varied outcomes from vitamin E are consistent with the differential functions of the isoforms of vitamin E. Mechanistic studies demonstrate that the vitamin E isoforms α-tocopherol and γ-tocopherol have opposite functions in regulation of allergic inflammation and development of allergic disease, with α-tocopherol having anti-inflammatory functions and γ-tocopherol having pro-inflammatory functions in allergy and asthma. Moreover, global differences in prevalence of asthma by country may be a result, at least in part, of differences in consumption of these two isoforms of tocopherols. It is critical in clinical and animal studies that measurements of the isoforms of tocopherols be determined in vehicles for the treatments, and in the plasma and/or tissues before and after intervention. As allergic inflammation is modifiable by tocopherol isoforms, differential regulation by tocopherol isoforms provide a foundation for development of interventions to improve lung function in disease and raise the possibility of early life dietary interventions to limit the development of lung disease.
Collapse
Affiliation(s)
- Joan M Cook-Mills
- Herman B Wells Center for Pediatric Research, Departments of Pediatrics and Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| | - Samantha H Averill
- Herman B Wells Center for Pediatric Research, Departments of Pediatrics and Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jacquelyn D Lajiness
- Herman B Wells Center for Pediatric Research, Departments of Pediatrics and Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| |
Collapse
|
7
|
Zhao L, Li Y, Xu T, Lv Q, Bi X, Liu X, Fu G, Zou Y, Ge J, Chen Z, Zhang W. Dendritic cell-mediated chronic low-grade inflammation is regulated by the RAGE-TLR4-PKCβ 1 signaling pathway in diabetic atherosclerosis. Mol Med 2022; 28:4. [PMID: 35062863 PMCID: PMC8780245 DOI: 10.1186/s10020-022-00431-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 01/03/2022] [Indexed: 11/16/2022] Open
Abstract
Background The unique mechanism of diabetic atherosclerosis has been a central research focus. Previous literature has reported that the inflammatory response mediated by dendritic cells (DCs) plays a vital role in the progression of atherosclerosis. The objective of the study was to explore the role of DCs in diabetes mellitus complicated by atherosclerosis. Methods ApoE−/− mice and bone marrow-derived DCs were used for in vivo and in vitro experiments, respectively. Masson’s staining and Oil-red-O staining were performed for atherosclerotic lesion assessment. The content of macrophages and DCs in plaque was visualized by immunohistochemistry. The expression of CD83 and CD86 were detected by flow cytometry. The fluctuations in the RNA levels of cytokines, chemokines, chemokine receptors and adhesions were analyzed by quantitative RT-PCR. The concentrations of IFN-γ and TNF-α were calculated using ELISA kits and the proteins were detected using western blot. Coimmunoprecipitation was used to detect protein–protein interactions. Results Compared with the ApoE−/− group, the volume of atherosclerotic plaques in the aortic root of diabetic ApoE−/− mice was significantly increased, numbers of macrophages and DCs were increased, and the collagen content in plaques decreased. The expression of CD83 and CD86 were significantly upregulated in splenic CD11c+ DCs derived from mice with hyperglycemia. Increased secretion of cytokines, chemokines, chemokine receptors, intercellular cell adhesion molecule (ICAM), and vascular cell adhesion molecule (VCAM) also were observed. The stimulation of advanced glycation end products plus oxidized low-density lipoprotein, in cultured BMDCs, further activated toll-like receptor 4, protein kinase C and receptor of AGEs, and induced immune maturation of DCs through the RAGE-TLR4-PKCβ1 signaling pathway that was bound together by intrinsic structures on the cell membrane. Administering LY333531 significantly increased the body weight of diabetic ApoE−/− mice, inhibited the immune maturation of spleen DCs, and reduced atherosclerotic plaques in diabetic ApoE−/− mice. Furthermore, the number of DCs and macrophages in atherosclerotic plaques was significantly reduced in the LY333531 group, and the collagen content was increased. Conclusions Diabetes mellitus aggravates chronic inflammation, and promotes atherosclerotic plaques in conjunction with hyperlipidemia, which at least in part through inducing the immune maturation of DCs, and its possible mechanism of action is through the RAGE-TLR4-pPKCβ1 signaling pathway. Supplementary Information The online version contains supplementary material available at 10.1186/s10020-022-00431-6.
Collapse
Affiliation(s)
- Liding Zhao
- Department of Cardiovascular Diseases, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, No 3 East of Qinchun Road, Hangzhou, Zhejiang, 310000, People's Republic of China.,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
| | - Ya Li
- Department of Cardiovascular Diseases, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, No 3 East of Qinchun Road, Hangzhou, Zhejiang, 310000, People's Republic of China.,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
| | - Tian Xu
- Department of Cardiovascular Diseases, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, No 3 East of Qinchun Road, Hangzhou, Zhejiang, 310000, People's Republic of China.,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
| | - Qingbo Lv
- Department of Cardiovascular Diseases, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, No 3 East of Qinchun Road, Hangzhou, Zhejiang, 310000, People's Republic of China.,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
| | - Xukun Bi
- Department of Cardiovascular Diseases, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, No 3 East of Qinchun Road, Hangzhou, Zhejiang, 310000, People's Republic of China.,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
| | - Xianglan Liu
- Department of Cardiovascular Diseases, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, No 3 East of Qinchun Road, Hangzhou, Zhejiang, 310000, People's Republic of China.,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
| | - Guosheng Fu
- Department of Cardiovascular Diseases, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, No 3 East of Qinchun Road, Hangzhou, Zhejiang, 310000, People's Republic of China.,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases of Zhongshan Hospital, Fudan University, Shanghai, China.,Institute of Biomedical Science, Fudan University, Shanghai, China
| | - Junbo Ge
- Shanghai Institute of Cardiovascular Diseases of Zhongshan Hospital, Fudan University, Shanghai, China.,Institute of Biomedical Science, Fudan University, Shanghai, China
| | - Zhaoyang Chen
- Heart Center of Fujian Province, Union Hospital, Fujian Medical University, 29 Xin-Quan Road, Fuzhou, 350001, People's Republic of China.
| | - Wenbin Zhang
- Department of Cardiovascular Diseases, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, No 3 East of Qinchun Road, Hangzhou, Zhejiang, 310000, People's Republic of China. .,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
8
|
Pachva MC, Lai H, Jia A, Rouleau M, Sorensen PH. Extracellular Vesicles in Reprogramming of the Ewing Sarcoma Tumor Microenvironment. Front Cell Dev Biol 2021; 9:726205. [PMID: 34604225 PMCID: PMC8484747 DOI: 10.3389/fcell.2021.726205] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/23/2021] [Indexed: 12/12/2022] Open
Abstract
Ewing sarcoma (EwS) is a highly aggressive cancer and the second most common malignant bone tumor of children and young adults. Although patients with localized disease have a survival rate of approximately 75%, the prognosis for patients with metastatic disease remains dismal (<30%) and has not improved in decades. Standard-of-care treatments include local therapies such as surgery and radiotherapy, in addition to poly-agent adjuvant chemotherapy, and are often associated with long-term disability and reduced quality of life. Novel targeted therapeutic strategies that are more efficacious and less toxic are therefore desperately needed, particularly for metastatic disease, given that the presence of metastasis remains the most powerful predictor of poor outcome in EwS. Intercellular communication within the tumor microenvironment is emerging as a crucial mechanism for cancer cells to establish immunosuppressive and cancer-permissive environments, potentially leading to metastasis. Altering this communication within the tumor microenvironment, thereby preventing the transfer of oncogenic signals and molecules, represents a highly promising therapeutic strategy. To achieve this, extracellular vesicles (EVs) offer a candidate mechanism as they are actively released by tumor cells and enriched with proteins and RNAs. EVs are membrane-bound particles released by normal and tumor cells, that play pivotal roles in intercellular communication, including cross-talk between tumor, stromal fibroblast, and immune cells in the local tumor microenvironment and systemic circulation. EwS EVs, including the smaller exosomes and larger microvesicles, have the potential to reprogram a diversity of cells in the tumor microenvironment, by transferring various biomolecules in a cell-specific manner. Insights into the various biomolecules packed in EwS EVs as cargos and the molecular changes they trigger in recipient cells of the tumor microenvironment will shed light on various potential targets for therapeutic intervention in EwS. This review details EwS EVs composition, their potential role in metastasis and in the reprogramming of various cells of the tumor microenvironment, and the potential for clinical intervention.
Collapse
Affiliation(s)
- Manideep C Pachva
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Horton Lai
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada.,Faculty of Science, University of British Columbia, Vancouver, BC, Canada
| | - Andy Jia
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada.,Faculty of Science, University of British Columbia, Vancouver, BC, Canada
| | - Melanie Rouleau
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Poul H Sorensen
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
9
|
Jin J, Li Y, Zhao Q, Chen Y, Fu S, Wu J. Coordinated regulation of immune contexture: crosstalk between STAT3 and immune cells during breast cancer progression. Cell Commun Signal 2021; 19:50. [PMID: 33957948 PMCID: PMC8101191 DOI: 10.1186/s12964-021-00705-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 01/11/2021] [Indexed: 12/24/2022] Open
Abstract
Recent insights into the molecular and cellular mechanisms underlying cancer development have revealed the tumor microenvironment (TME) immune cells to functionally affect the development and progression of breast cancer. However, insufficient evidence of TME immune modulators limit the clinical application of immunotherapy for advanced and metastatic breast cancers. Intercellular STAT3 activation of immune cells plays a central role in breast cancer TME immunosuppression and distant metastasis. Accumulating evidence suggests that targeting STAT3 and/or in combination with radiotherapy may enhance anti-cancer immune responses and rescue the systemic immunologic microenvironment in breast cancer. Indeed, apart from its oncogenic role in tumor cells, the functions of STAT3 in TME of breast cancer involve multiple types of immunosuppression and is associated with tumor cell metastasis. In this review, we summarize the available information on the functions of STAT3-related immune cells in TME of breast cancer, as well as the specific upstream and downstream targets. Additionally, we provide insights about the potential immunosuppression mechanisms of each type of evaluated immune cells. Video abstract.
Collapse
Affiliation(s)
- Jing Jin
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| | - Yi Li
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| | - Qijie Zhao
- Department of Radiologic Technology, Center of Excellence for Molecular Imaging (CEMI), Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200 Thailand
- Department of Pathophysiology, College of Basic Medical Science, Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| | - Yue Chen
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, 646000 Sichuan People’s Republic of China
- Academician (Expert) Workstation of Sichuan Province, Luzhou, 646000 Sichuan People’s Republic of China
| | - Shaozhi Fu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| | - JingBo Wu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, 646000 Sichuan People’s Republic of China
- Academician (Expert) Workstation of Sichuan Province, Luzhou, 646000 Sichuan People’s Republic of China
| |
Collapse
|
10
|
Signal Transduction in Immune Cells and Protein Kinases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1275:133-149. [PMID: 33539014 DOI: 10.1007/978-3-030-49844-3_5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Immune response relies upon several intracellular signaling events. Among the protein kinases involved in these pathways, members of the protein kinase C (PKC) family are prominent molecules because they have the capacity to acutely and reversibly modulate effector protein functions, controlling both spatial distribution and dynamic properties of the signals. Different PKC isoforms are involved in distinct signaling pathways, with selective functions in a cell-specific manner.In innate system, Toll-like receptor signaling is the main molecular event triggering effector functions. Various isoforms of PKC can be common to different TLRs, while some of them are specific for a certain type of TLR. Protein kinases involvement in innate immune cells are presented within the chapter emphasizing their coordination in many aspects of immune cell function and, as important players in immune regulation.In adaptive immunity T-cell receptor and B-cell receptor signaling are the main intracellular pathways involved in seminal immune specific cellular events. Activation through TCR and BCR can have common intracellular pathways while others can be specific for the type of receptor involved or for the specific function triggered. Various PKC isoforms involvement in TCR and BCR Intracellular signaling will be presented as positive and negative regulators of the immune response events triggered in adaptive immunity.
Collapse
|
11
|
Galbiati V, Marinovich M, Corsini E. Mechanistic understanding of dendritic cell activation in skin sensitization: additional evidences to support potency classification. Toxicol Lett 2020; 322:50-57. [DOI: 10.1016/j.toxlet.2020.01.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 01/14/2020] [Accepted: 01/16/2020] [Indexed: 01/23/2023]
|
12
|
Lalor R, O'Neill S. Bovine κ-casein induces a hypo-responsive DC population which exhibit a reduced capacity to elicit T-cell responses. J Funct Foods 2020. [DOI: 10.1016/j.jff.2019.103620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
13
|
Pizzino G, Irrera N, Cucinotta M, Pallio G, Mannino F, Arcoraci V, Squadrito F, Altavilla D, Bitto A. Oxidative Stress: Harms and Benefits for Human Health. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:8416763. [PMID: 28819546 PMCID: PMC5551541 DOI: 10.1155/2017/8416763] [Citation(s) in RCA: 2261] [Impact Index Per Article: 282.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 07/05/2017] [Indexed: 02/07/2023]
Abstract
Oxidative stress is a phenomenon caused by an imbalance between production and accumulation of oxygen reactive species (ROS) in cells and tissues and the ability of a biological system to detoxify these reactive products. ROS can play, and in fact they do it, several physiological roles (i.e., cell signaling), and they are normally generated as by-products of oxygen metabolism; despite this, environmental stressors (i.e., UV, ionizing radiations, pollutants, and heavy metals) and xenobiotics (i.e., antiblastic drugs) contribute to greatly increase ROS production, therefore causing the imbalance that leads to cell and tissue damage (oxidative stress). Several antioxidants have been exploited in recent years for their actual or supposed beneficial effect against oxidative stress, such as vitamin E, flavonoids, and polyphenols. While we tend to describe oxidative stress just as harmful for human body, it is true as well that it is exploited as a therapeutic approach to treat clinical conditions such as cancer, with a certain degree of clinical success. In this review, we will describe the most recent findings in the oxidative stress field, highlighting both its bad and good sides for human health.
Collapse
Affiliation(s)
- Gabriele Pizzino
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Natasha Irrera
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Mariapaola Cucinotta
- Department of Biomedical Sciences, Dentistry and Morphological and Functional Images, University of Messina, Messina, Italy
| | - Giovanni Pallio
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Federica Mannino
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Vincenzo Arcoraci
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Francesco Squadrito
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Domenica Altavilla
- Department of Biomedical Sciences, Dentistry and Morphological and Functional Images, University of Messina, Messina, Italy
| | - Alessandra Bitto
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| |
Collapse
|
14
|
Transcriptional mechanism of vascular endothelial growth factor-induced expression of protein kinase CβII in chronic lymphocytic leukaemia cells. Sci Rep 2017; 7:43228. [PMID: 28233872 PMCID: PMC5324130 DOI: 10.1038/srep43228] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 01/20/2017] [Indexed: 12/24/2022] Open
Abstract
A key feature of chronic lymphocytic leukaemia (CLL) cells is overexpressed protein kinase CβII (PKCβII), an S/T kinase important in the pathogenesis of this and other B cell malignancies. The mechanisms contributing to enhanced transcription of the gene coding for PKCβII, PRKCB, in CLL cells remain poorly described, but could be important because of potential insight into how the phenotype of these cells is regulated. Here, we show that SP1 is the major driver of PKCβII expression in CLL cells where enhanced association of this transcription factor with the PRKCB promoter is likely because of the presence of histone marks permissive of gene activation. We also show how vascular endothelial growth factor (VEGF) regulates PRKCB promoter function in CLL cells, stimulating PKCβ gene transcription via increased association of SP1 and decreased association of STAT3. Taken together, these results are the first to demonstrate a clear role for SP1 in the up regulation of PKCβII expression in CLL cells, and the first to link SP1 with the pathogenesis of this and potentially other B cell malignancies where PKCβII is overexpressed.
Collapse
|
15
|
Galli F, Azzi A, Birringer M, Cook-Mills JM, Eggersdorfer M, Frank J, Cruciani G, Lorkowski S, Özer NK. Vitamin E: Emerging aspects and new directions. Free Radic Biol Med 2017; 102:16-36. [PMID: 27816611 DOI: 10.1016/j.freeradbiomed.2016.09.017] [Citation(s) in RCA: 267] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 09/11/2016] [Accepted: 09/22/2016] [Indexed: 12/30/2022]
Abstract
The discovery of vitamin E will have its 100th anniversary in 2022, but we still have more questions than answers regarding the biological functions and the essentiality of vitamin E for human health. Discovered as a factor essential for rat fertility and soon after characterized for its properties of fat-soluble antioxidant, vitamin E was identified to have signaling and gene regulation effects in the 1980s. In the same years the cytochrome P-450 dependent metabolism of vitamin E was characterized and a first series of studies on short-chain carboxyethyl metabolites in the 1990s paved the way to the hypothesis of a biological role for this metabolism alternative to vitamin E catabolism. In the last decade other physiological metabolites of vitamin E have been identified, such as α-tocopheryl phosphate and the long-chain metabolites formed by the ω-hydroxylase activity of cytochrome P-450. Recent findings are consistent with gene regulation and homeostatic roles of these metabolites in different experimental models, such as inflammatory, neuronal and hepatic cells, and in vivo in animal models of acute inflammation. Molecular mechanisms underlying these responses are under investigation in several laboratories and side-glances to research on other fat soluble vitamins may help to move faster in this direction. Other emerging aspects presented in this review paper include novel insights on the mechanisms of reduction of the cardiovascular risk, immunomodulation and antiallergic effects, neuroprotection properties in models of glutamate excitotoxicity and spino-cerebellar damage, hepatoprotection and prevention of liver toxicity by different causes and even therapeutic applications in non-alcoholic steatohepatitis. We here discuss these topics with the aim of stimulating the interest of the scientific community and further research activities that may help to celebrate this anniversary of vitamin E with an in-depth knowledge of its action as vitamin.
Collapse
Affiliation(s)
- Francesco Galli
- Department of Pharmaceutical Sciences, University of Perugia, Laboratory of Clinical Biochemistry and Nutrition, Via del Giochetto, 06126 Perugia, Italy.
| | - Angelo Azzi
- USDA-HNRCA at Tufts University, 711 Washington St., Boston, MA 02111, United States.
| | - Marc Birringer
- Department of Nutritional, Food and Consumer Sciences, Fulda University of Applied Sciences, Leipziger Straße 123, 36037 Fulda, Germany.
| | - Joan M Cook-Mills
- Allergy/Immunology Division, Northwestern University, 240 E Huron, Chicago, IL 60611, United States.
| | | | - Jan Frank
- Institute of Biological Chemistry and Nutrition, University of Hohenheim, Garbenstr. 28, 70599 Stuttgart, Germany.
| | - Gabriele Cruciani
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Italy.
| | - Stefan Lorkowski
- Institute of Nutrition, Friedrich Schiller University Jena, Dornburger Str. 25, 07743 Jena, Germany; Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Germany.
| | - Nesrin Kartal Özer
- Department of Biochemistry, Faculty of Medicine, Genetic and Metabolic Diseases Research Center (GEMHAM), Marmara University, 34854 Maltepe, Istanbul, Turkey.
| |
Collapse
|
16
|
Abstract
Heroin addiction is a complex psychiatric disorder with a chronic course and a high relapse rate, which results from the interaction between genetic and environmental factors. Heroin addiction has a substantial heritability in its etiology; hence, identification of individuals with a high genetic propensity to heroin addiction may help prevent the occurrence and relapse of heroin addiction and its complications. The study aimed to identify a small set of genetic signatures that may reliably predict the individuals with a high genetic propensity to heroin addiction. We first measured the transcript level of 13 genes (RASA1, PRKCB, PDK1, JUN, CEBPG, CD74, CEBPB, AUTS2, ENO2, IMPDH2, HAT1, MBD1, and RGS3) in lymphoblastoid cell lines in a sample of 124 male heroin addicts and 124 male control subjects using real-time quantitative PCR. Seven genes (PRKCB, PDK1, JUN, CEBPG, CEBPB, ENO2, and HAT1) showed significant differential expression between the 2 groups. Further analysis using 3 statistical methods including logistic regression analysis, support vector machine learning analysis, and a computer software BIASLESS revealed that a set of 4 genes (JUN, CEBPB, PRKCB, ENO2, or CEBPG) could predict the diagnosis of heroin addiction with the accuracy rate around 85% in our dataset. Our findings support the idea that it is possible to identify genetic signatures of heroin addiction using a small set of expressed genes. However, the study can only be considered as a proof-of-concept study. As the establishment of lymphoblastoid cell line is a laborious and lengthy process, it would be more practical in clinical settings to identify genetic signatures for heroin addiction directly from peripheral blood cells in the future study.
Collapse
Affiliation(s)
- Shaw-Ji Chen
- Institute of Medical Sciences, Tzu Chi University, Hualien
- Department of Psychiatry, Mackay Memorial Hospital, Taitung Branch
| | - Ding-Lieh Liao
- Department of Health Executive Yuan, Bali Psychiatric Center
| | - Tsu-Wang Shen
- Institute of Medical Sciences, Tzu Chi University, Hualien
| | - Hsin-Chou Yang
- Institute of Statistical Science, Academia Sinica, Taipei
| | - Kuang-Chi Chen
- Institute of Medical Sciences, Tzu Chi University, Hualien
| | - Chia-Hsiang Chen
- Department of Psychiatry, Chang Gung Memorial Hospital at Linkou
- Department and Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
- Correspondence: Chia-Hsiang Chen, Department of Psychiatry, Chang Gung Memorial Hospital at Linkou, No. 5 Fusing Street, Kueishan, Taoyuan, 333 Taiwan (e-mail: )
| |
Collapse
|
17
|
Zymosan and PMA activate the immune responses of Mutz3-derived dendritic cells synergistically. Immunol Lett 2015; 167:41-6. [PMID: 26183538 DOI: 10.1016/j.imlet.2015.07.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 06/10/2015] [Accepted: 07/06/2015] [Indexed: 01/22/2023]
Abstract
Beta-glucan (β-glucan) including zymosan has been known as a super food because of its multifunctional activities, such as the enhancement of immune responses. To study the functional mechanism of β-glucan in immune stimulation, the effect of zymosan on dendritic cell (DC) was investigated by monitoring the production of TNF-α, a pro-inflammatory cytokine. DC was differentiated from Mutz-3, a human acute myeloid leukemia cell line, by cytokine treatment and characterized. DC-specific cell surface markers were increased during the differentiation. Especially, Dectin-1, a β-glucan receptor, was upregulated during DC differentiation, and mediated zymosan-induced TNF-α production, which was inhibited by silencing of dectin-1. Zymosan exhibited synergistic effect with other immune stimuli such as lipopolysaccharide (LPS) and phorbol 12-myristate 13-acetate (PMA), a well-known PKC activator. Simultaneous treatment of zymosan and PMA enhanced the nuclear translocation of NF-κB subunits, p50 and p65, mediating the increase of TNF-α production. Bay 11-7082, an NF-κB inhibitor, blocked morphological changes and TNF-α production induced by zymosan and/or PMA treatment. Western blot analysis has showed zymosan-Dectin-1 pathway mediated destructive phosphorylation of inhibitor of NF-κB (IκB) kinase α subunit (IKKα) in IKK complexes, while PMA-PKC pathway regulated selective phosphorylation and degradation of IKKβ. Simultaneous phosphorylation of separate IKK subunits by co-treatment of zymosan and PMA resulted in cooperative activation of NF-κB and TNF-α production.
Collapse
|
18
|
Corsini E, Galbiati V, Pinto A, Davin A, Polito L, Guaita A, Racchi M. Immunostimulatory effects of RACK1 pseudosubstrate in human leukocytes obtained from young and old donors. Oncotarget 2015; 6:6524-34. [PMID: 25779661 PMCID: PMC4466631 DOI: 10.18632/oncotarget.3002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 12/21/2014] [Indexed: 11/25/2022] Open
Abstract
Aims of this study were to investigate the ability of RACK1 pseudosubstrate alone or in combination with classical immune stimuli to activate human leukocytes, and to restore age-associated immune defects.A total of 25 donors (17 old donors, 77-79 yrs; 8 young donors, 25-34 yrs) were enrolled. To evaluate the effect of RACK1 pseudosubstrate on cytokine production and CD86 expression the whole blood assay was used. Cultures were treated with RACK1 pseudosubstrate in the presence or absence of lipopolysaccharide (LPS) or phytohaemagglutinin (PHA) and incubated for 24 h or 48 h for LPS-induced CD86 expression, TNF-α, IL-6, IL-8, IL-10 production, and PHA-induced IL-4, IL-10, IFN-γ, respectively. RACK1 pseudosubstrate alone induced IL-6, IL-8, and CD86 expression in both young and old donors, and IFN-γ in old donors. In combination with LPS an increase in IL-8, IL-10 and TNF-α was observed, also resulting in restoration of age-associated defective production, while no changes in the other parameters investigated were found.Even if based on a small sample size, these results suggest the possibility to by-pass some of age-associated immune alterations, which may be beneficial in situations were natural immune stimulation is required, and highlight a different role of PKCβ in immune cells activation.
Collapse
Affiliation(s)
- Emanuela Corsini
- Laboratory of Toxicology, DiSFeB, Università degli Studi di Milano, Milan, Italy
| | - Valentina Galbiati
- Laboratory of Toxicology, DiSFeB, Università degli Studi di Milano, Milan, Italy
| | - Antonella Pinto
- Department of Drug Sciences - Pharmacology, University of Pavia, Pavia, Italy
| | | | | | | | - Marco Racchi
- Department of Drug Sciences - Pharmacology, University of Pavia, Pavia, Italy
| |
Collapse
|
19
|
Farren MR, Carlson LM, Netherby CS, Lindner I, Li PK, Gabrilovich DI, Abrams SI, Lee KP. Tumor-induced STAT3 signaling in myeloid cells impairs dendritic cell generation by decreasing PKCβII abundance. Sci Signal 2014; 7:ra16. [PMID: 24550541 DOI: 10.1126/scisignal.2004656] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
A major mechanism by which cancers escape control by the immune system is by blocking the differentiation of myeloid cells into dendritic cells (DCs), immunostimulatory cells that activate antitumor T cells. Tumor-dependent activation of signal transducer and activator of transcription 3 (STAT3) signaling in myeloid progenitor cells is thought to cause this block in their differentiation. In addition, a signaling pathway through protein kinase C βII (PKCβII) is essential for the differentiation of myeloid cells into DCs. We found in humans and mice that breast cancer cells substantially decreased the abundance of PKCβII in myeloid progenitor cells through a mechanism involving the enhanced activation of STAT3 signaling by soluble, tumor-derived factors (TDFs). STAT3 bound to previously undescribed negative regulatory elements within the promoter of PRKCB, which encodes PKCβII. We also found a previously undescribed counter-regulatory mechanism through which the activity of PKCβII inhibited tumor-dependent STAT3 signaling by decreasing the abundance of cell surface receptors, such as cytokine and growth factor receptors, that are activated by TDFs. Together, these data suggest that a previously unrecognized cross-talk mechanism between the STAT3 and PKCβII signaling pathways provides the molecular basis for the tumor-induced blockade in the differentiation of myeloid cells, and suggest that enhancing PKCβII activity may be a therapeutic strategy to alleviate cancer-mediated suppression of the immune system.
Collapse
Affiliation(s)
- Matthew R Farren
- 1Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Koorella C, Nair JR, Murray ME, Carlson LM, Watkins SK, Lee KP. Novel regulation of CD80/CD86-induced phosphatidylinositol 3-kinase signaling by NOTCH1 protein in interleukin-6 and indoleamine 2,3-dioxygenase production by dendritic cells. J Biol Chem 2014; 289:7747-62. [PMID: 24415757 DOI: 10.1074/jbc.m113.519686] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Dendritic cells (DC) play a critical role in modulating antigen-specific immune responses elicited by T cells via engagement of the prototypic T cell costimulatory receptor CD28 by the cognate ligands CD80/CD86, expressed on DC. Although CD28 signaling in T cell activation has been well characterized, it has only recently been shown that CD80/CD86, which have no demonstrated binding domains for signaling proteins in their cytoplasmic tails, nonetheless also transduce signals to the DC. Functionally, CD80/CD86 engagement results in DC production of the pro-inflammatory cytokine IL-6, which is necessary for full T cell activation. However, ligation of CD80/CD86 by CTLA4 also induces DC production of the immunosuppressive enzyme indoleamine 2,3-dioxygenase (IDO), which depletes local pools of the essential amino acid tryptophan, resulting in blockade of T cell activation. Despite the significant role of CD80/CD86 in immunological processes and the seemingly opposing roles they play by producing IL-6 and IDO upon their activation, how CD80/CD86 signal remains poorly understood. We have now found that cross-linking CD80/CD86 in human DC activates the PI3K/AKT pathway. This results in phosphorylation/inactivation of its downstream target, FOXO3A, and alleviates FOXO3A-mediated suppression of IL-6 expression. A second event downstream of AKT phosphorylation is activation of the canonical NF-κB pathway, which induces IL-6 expression. In addition to these downstream pathways, we unexpectedly found that CD80/CD86-induced PI3K signaling is regulated by previously unrecognized cross-talk with NOTCH1 signaling. This cross-talk is facilitated by NOTCH-mediated up-regulation of the expression of prolyl isomerase PIN1, which in turn increases enzyme activity of casein kinase II. Subsequently, phosphatase and tensin homolog (which suppresses PI3K activity) is inactivated via phosphorylation by casein kinase II. This results in full activation of PI3K signaling upon cross-linking CD80/CD86. Similar to IL-6, we have found that CD80/CD86-induced IDO production by DC at late time points is also dependent upon the PI3K → AKT → NF-κB pathway and requires cross-talk with NOTCH signaling. These data further suggest that the same signaling pathways downstream of DC CD80/CD86 cross-linking induce early IL-6 production to enhance T cell activation, followed by later IDO production to self-limit this activation. In addition to characterizing the pathways downstream of CD80/CD86 in IL-6 and IDO production, identification of a novel cross-talk between NOTCH1 and PI3K signaling may provide new insights in other biological processes where PI3K signaling plays a major role.
Collapse
Affiliation(s)
- Chandana Koorella
- From the Department of Immunology, Roswell Park Cancer Institute, Buffalo, New York 14263 and
| | | | | | | | | | | |
Collapse
|
21
|
Bade-Döding C, Göttmann W, Baigger A, Farren M, Lee KP, Blasczyk R, Huyton T. Autocrine GM-CSF transcription in the leukemic progenitor cell line KG1a is mediated by the transcription factor ETS1 and is negatively regulated through SECTM1 mediated ligation of CD7. Biochim Biophys Acta Gen Subj 2013; 1840:1004-13. [PMID: 24211252 DOI: 10.1016/j.bbagen.2013.10.043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Revised: 09/17/2013] [Accepted: 10/27/2013] [Indexed: 11/17/2022]
Abstract
BACKGROUND CD7 expression is found on ~30% of acute myeloblastic leukemias (AML). The leukemic progenitor cell line KG1a (CD7+) constitutively expresses GM-CSF while the parental KG1 (CD7-) cell line does not. This study focuses on the molecular basis of CD7 mediated GM-CSF regulation. METHODS KG1a cells were treated with recombinant SECTM1-Fc protein, the PI3K kinase inhibitors wortmannin, LY292004, or PI4K activator spermine. Stable KG1-CD7+, KG1a-shCD7, KG1a-shETS1 as well as KG1a-GFP, KG1a-PKCβII-GFP cell lines were generated and the levels of CD7, GM-CSF and ETS-1 mRNA and protein were compared by real-time-PCR, western blotting, flow cytometry and ELISA. RESULTS SECTM1 is expressed in Human Bone Marrow Endothelial Cells (HBMEC) and its expression can be upregulated by both IFN-γ. KG1a cells demonstrated high expression levels of CD7 and ETS-1 allowing a constitutative signaling through the PI3K/Atk pathway to promote GM-CSF expression, while KG1 cells with low expression of CD7 and ETS-1 showed low GM-CSF expression. On KG1a cells GM-CSF expression could be negatively regulated by PI3K inhibitors or by recombinant SECTM1-Fc. Overexpression of CD7 in KG1 cells was insufficient to promote GM-CSF expression, while silencing of CD7 or ETS-1 resulted in reduced GM-CSF expression levels. Differentiation capable KG1a cells overexpressing PKCβII illustrated complete loss of CD7, but maintained normal levels of both ETS-1 and GM-CSF expression. CONCLUSION These findings add an additional layer to the previously described autocrine/paracrine signaling between leukemic progenitor cells and the bone marrow microenvironment and highlight a role for SECTM1 in both normal and malignant hematopoiesis. GENERAL SIGNIFICANCE This work shows that SECTM1 secreted from bone marrow stromal cells may interact with CD7 to influence GM-CSF expression in leukemic cells.
Collapse
Affiliation(s)
- Christina Bade-Döding
- Institute for Transfusion Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - Wiebke Göttmann
- Institute for Transfusion Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - Anja Baigger
- Institute for Transfusion Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - Matthew Farren
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14226, USA
| | - Kelvin P Lee
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14226, USA
| | - Rainer Blasczyk
- Institute for Transfusion Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - Trevor Huyton
- Institute for Transfusion Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany.
| |
Collapse
|
22
|
Corsini E, Galbiati V, Esser PR, Pinto A, Racchi M, Marinovich M, Martin SF, Galli CL. Role of PKC-β in chemical allergen-induced CD86 expression and IL-8 release in THP-1 cells. Arch Toxicol 2013; 88:415-24. [PMID: 24136171 DOI: 10.1007/s00204-013-1144-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 10/07/2013] [Indexed: 10/26/2022]
Abstract
We previously demonstrated an age-related decrease in receptor for activated C-kinase (RACK-1) expression and functional deficit in Langerhans cells' responsiveness. This defect specifically involves the translocation of protein kinase C (PKC)-β. The purpose of this study was to investigate the role of RACK-1 and PKC-β in chemical allergen-induced CD86 expression and IL-8 release in the human promyelocytic cell line THP-1 and primary human dendritic cells (DC). Dinitrochlorobenzene, p-phenylenediamine and diethyl maleate were used as contact allergens. The selective cell-permeable inhibitor of PKC-β and the broad PKC inhibitor GF109203X completely prevented chemical allergen- or lipopolysaccharide (LPS)-induced CD86 expression and significantly modulated IL-8 release (50 % reduction). The selective cell-permeable inhibitor of PKC-ε (also known to bind to RACK-1) failed to modulate allergen- or LPS-induced CD86 expression or allergen-induced IL-8 release, while modulating LPS-induced IL-8 release. The use of a RACK-1 pseudosubstrate, which directly activates PKC-β, resulted in dose-related increase in CD86 expression and IL-8 release. Similar results were obtained with human DC, confirming the relevance of results obtained in THP-1 cells. Overall, our findings demonstrate the role of PKC-β and RACK-1 in allergen-induced CD86 expression and IL-8 production, supporting a central role of PKC-β in the initiation of chemical allergen-induced DC activation.
Collapse
Affiliation(s)
- Emanuela Corsini
- Laboratory of Toxicology, Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Via G. Balzaretti 9, 20133, Milan, Italy,
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Matsumoto T, Hasegawa H, Onishi S, Ishizaki J, Suemori K, Yasukawa M. Protein kinase C inhibitor generates stable human tolerogenic dendritic cells. THE JOURNAL OF IMMUNOLOGY 2013; 191:2247-57. [PMID: 23878315 DOI: 10.4049/jimmunol.1203053] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Tolerogenic dendritic cells (DCs) are a promising tool for a specific form of cellular therapy whereby immunological tolerance can be induced in the context of transplantation and autoimmunity. From libraries of bioactive lipids, nuclear receptor ligands, and kinase inhibitors, we screened conventional protein kinase C inhibitors (PKCIs) bisindolylmaleimide I, Gö6983, and Ro32-0432 with strong tolerogenic potential. PKCI-treated human DCs were generated by subjecting them to a maturation process after differentiation of immature DCs. The PKCI-treated DCs had a semimature phenotype, showing high production of IL-10, and efficiently induced IL-10-producing T cells and functional Foxp3(+) regulatory T cells from naive CD4(+) T cells, thus eliciting a strong immunosuppressive function. They also showed CCR7 expression and sufficient capacity for migration toward CCR7 ligands. Additionally, PKCI-treated DCs were highly stable when exposed to inflammatory stimuli such as proinflammatory cytokines or LPS. Conventional PKCIs inhibited NF-κB activation of both the canonical and noncanonical pathways of DC maturation, thus suppressing the expression of costimulatory molecules and IL-12 production. High production of IL-10 in PKCI-treated DCs was due to not only an increase of intracellular cAMP, but also a synergistic effect of increased cAMP and NF-κB inhibition. Moreover, PKCI-treated mouse DCs that had properties similar to PKCI-treated human DCs prevented graft-versus-host disease in a murine model of acute graft-versus-host disease. Conventional PKCI-treated DCs may be useful for tolerance-inducing therapy, as they satisfy the required functional characteristics for clinical-grade tolerogenic DCs.
Collapse
Affiliation(s)
- Takuya Matsumoto
- Department of Bioregulatory Medicine, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan
| | | | | | | | | | | |
Collapse
|
24
|
Huemer HP, Geiger M, Posch W, Krumböck N, Fresser F, Wilflingseder D, Uberall F. Protein kinase C overexpression does not enhance immune-stimulatory surface markers of vaccinia-infected dendritic cells and DC cell lines. Immunol Invest 2013; 42:164-77. [PMID: 23323525 PMCID: PMC9491103 DOI: 10.3109/08820139.2012.750340] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
One of the shortcomings of vaccinia virus (VACV) as immunization vector is the down-regulation of HLA and costimulatory molecules in antigen presenting cells. To overcome this problem we investigated the use of protein kinase C (PKC) as immune stimulatory agent. Thus several classical and atypical PKCs were inserted into wild-type or attenuated VACV using recombination into the hemagglutinin gene and the expression driven by the VACV 7,5K-IE gene promoter. Recombinant constructs expressing PKC-alpha, -beta, -theta as well as wild-type, constitutive active or dominant negative PKC-zeta constructs were generated. Additional constructs expressing PKB/Akt1 and ICAM-1 were used for comparison. Immature and mature peripheral blood derived-dendritic cells (DC) as well as lymphoid cell lines capable of obtaining a DC-like phenotype upon mitogen stimulation were infected. Disappointingly, VACV-driven PKC overexpression did not significantly enhance expression of various activation markers or costimulatory molecules tested. Neither CD86 nor HLA-DR expression was upregulated and also no influence on the maturation of DC, as measured by DC-SIGN and CD83, was observed. However, VACV did not interfere with LPS induced up-regulation of CD83 and did not lead to substantial apoptosis of infected DC within the first 24 hours.
Collapse
Affiliation(s)
- Hartwig P Huemer
- Department of Hygiene, Microbiology & Social Medicine, Medical University Innsbruck, Innsbruck, Austria.
| | | | | | | | | | | | | |
Collapse
|
25
|
Black AR, Black JD. Protein kinase C signaling and cell cycle regulation. Front Immunol 2013; 3:423. [PMID: 23335926 PMCID: PMC3547298 DOI: 10.3389/fimmu.2012.00423] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Accepted: 12/21/2012] [Indexed: 12/20/2022] Open
Abstract
A link between T cell proliferation and the protein kinase C (PKC) family of serine/threonine kinases has been recognized for about 30 years. However, despite the wealth of information on PKC-mediated control of, T cell activation, understanding of the effects of PKCs on the cell cycle machinery in this cell type remains limited. Studies in other systems have revealed important cell cycle-specific effects of PKC signaling that can either positively or negatively impact proliferation. The outcome of PKC activation is highly context-dependent, with the precise cell cycle target(s) and overall effects determined by the specific isozyme involved, the timing of PKC activation, the cell type, and the signaling environment. Although PKCs can regulate all stages of the cell cycle, they appear to predominantly affect G0/G1 and G2. PKCs can modulate multiple cell cycle regulatory molecules, including cyclins, cyclin-dependent kinases (cdks), cdk inhibitors and cdc25 phosphatases; however, evidence points to Cip/Kip cdk inhibitors and D-type cyclins as key mediators of PKC-regulated cell cycle-specific effects. Several PKC isozymes can target Cip/Kip proteins to control G0/G1 → S and/or G2 → M transit, while effects on D-type cyclins regulate entry into and progression through G1. Analysis of PKC signaling in T cells has largely focused on its roles in T cell activation; thus, observed cell cycle effects are mainly positive. A prominent role is emerging for PKCθ, with non-redundant functions of other isozymes also described. Additional evidence points to PKCδ as a negative regulator of the cell cycle in these cells. As in other cell types, context-dependent effects of individual isozymes have been noted in T cells, and Cip/Kip cdk inhibitors and D-type cyclins appear to be major PKC targets. Future studies are anticipated to take advantage of the similarities between these various systems to enhance understanding of PKC-mediated cell cycle regulation in T cells.
Collapse
Affiliation(s)
- Adrian R Black
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center Omaha, NE, USA
| | | |
Collapse
|
26
|
Racke FK, Baird M, Barth RF, Huo T, Yang W, Gupta N, Weldon M, Rutledge H. Unique in vitro and in vivo thrombopoietic activities of ingenol 3,20 dibenzoate, a Ca(++)-independent protein kinase C isoform agonist. PLoS One 2012; 7:e51059. [PMID: 23284657 PMCID: PMC3528756 DOI: 10.1371/journal.pone.0051059] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Accepted: 10/29/2012] [Indexed: 11/19/2022] Open
Abstract
Thrombopoiesis following severe bone marrow injury frequently is delayed, thereby resulting in life-threatening thrombocytopenia for which there are limited treatment options. The reasons for these delays in recovery are not well understood. Protein kinase C (PKC) agonists promote megakaryocyte differentiation in leukemia cell lines and primary cells. However, little is known about the megakaryopoietic effects of PKC agonists on primary CD34+ cells grown in culture or in vivo. Here we present evidence that the novel PKC isoform-selective agonist 3,20 ingenol dibenzoate (IDB) potently stimulates early megakaryopoiesis of human CD34+ cells. In contrast, broad spectrum PKC agonists failed to do so. In vivo, a single intraperitoneal injection of IDB selectively increased platelets in mice without affecting hemoglobin or white counts. Finally, IDB strongly mitigated radiation-induced thrombocytopenia, even when administered 24 hours after irradiation. Our data demonstrate that novel PKC isoform agonists such as IDB may represent a unique therapeutic strategy for accelerating the recovery of platelet counts following severe marrow injury.
Collapse
Affiliation(s)
- Frederick K Racke
- Department of Pathology, The Ohio State University School of Medicine, Columbus, Ohio, United States of America.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Nair JR, Carlson LM, Koorella C, Rozanski CH, Byrne GE, Bergsagel PL, Shaughnessy JP, Boise LH, Chanan-Khan A, Lee KP. CD28 expressed on malignant plasma cells induces a prosurvival and immunosuppressive microenvironment. THE JOURNAL OF IMMUNOLOGY 2011; 187:1243-53. [PMID: 21715687 DOI: 10.4049/jimmunol.1100016] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Interactions between the malignant plasma cells of multiple myeloma and stromal cells within the bone marrow microenvironment are essential for myeloma cell survival, mirroring the same dependence of normal bone marrow-resident long-lived plasma cells on specific marrow niches. These interactions directly transduce prosurvival signals to the myeloma cells and also induce niche production of supportive soluble factors. However, despite their central importance, the specific molecular and cellular components involved remain poorly characterized. We now report that the prototypic T cell costimulatory receptor CD28 is overexpressed on myeloma cells during disease progression and in the poor-prognosis subgroups and plays a previously unrecognized role as a two-way molecular bridge to support myeloid stromal cells in the microenvironment. Engagement by CD28 to its ligand CD80/CD86 on stromal dendritic cell directly transduces a prosurvival signal to myeloma cell, protecting it against chemotherapy and growth factor withdrawal-induced death. Simultaneously, CD28-mediated ligation of CD80/CD86 induces the stromal dendritic cell to produce the prosurvival cytokine IL-6 (involving novel cross-talk with the Notch pathway) and the immunosuppressive enzyme IDO. These findings identify CD28 and CD80/CD86 as important molecular components of the interaction between myeloma cells and the bone marrow microenvironment, point to similar interaction for normal plasma cells, and suggest novel therapeutic strategies to target malignant and pathogenic (e.g., in allergy and autoimmunity) plasma cells.
Collapse
Affiliation(s)
- Jayakumar R Nair
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14226, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Gambone JE, Dusaban SS, Loperena R, Nakata Y, Shetzline SE. The c-Myb target gene neuromedin U functions as a novel cofactor during the early stages of erythropoiesis. Blood 2011; 117:5733-43. [PMID: 21378276 PMCID: PMC3110030 DOI: 10.1182/blood-2009-09-242131] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2009] [Accepted: 02/02/2011] [Indexed: 11/20/2022] Open
Abstract
The requirement of c-Myb during erythropoiesis spurred an interest in identifying c-Myb target genes that are important for erythroid development. Here, we determined that the neuropeptide neuromedin U (NmU) is a c-Myb target gene. Silencing NmU, c-myb, or NmU's cognate receptor NMUR1 expression in human CD34(+) cells impaired burst-forming unit-erythroid (BFU-E) and colony-forming unit-erythroid (CFU-E) formation compared with control. Exogenous addition of NmU peptide to NmU or c-myb siRNA-treated CD34(+) cells rescued BFU-E and yielded a greater number of CFU-E than observed with control. No rescue of BFU-E and CFU-E growth was observed when NmU peptide was exogenously added to NMUR1 siRNA-treated cells compared with NMUR1 siRNA-treated cells cultured without NmU peptide. In K562 and CD34(+) cells, NmU activated protein kinase C-βII, a factor associated with hematopoietic differentiation-proliferation. CD34(+) cells cultured under erythroid-inducing conditions, with NmU peptide and erythropoietin added at day 6, revealed an increase in endogenous NmU and c-myb gene expression at day 8 and a 16% expansion of early erythroblasts at day 10 compared to cultures without NmU peptide. Combined, these data strongly support that the c-Myb target gene NmU functions as a novel cofactor for erythropoiesis and expands early erythroblasts.
Collapse
Affiliation(s)
- Julia E Gambone
- Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | | | | | | | | |
Collapse
|
29
|
Farren MR, Carlson LM, Lee KP. Tumor-mediated inhibition of dendritic cell differentiation is mediated by down regulation of protein kinase C beta II expression. Immunol Res 2010; 46:165-76. [PMID: 19756409 DOI: 10.1007/s12026-009-8118-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Tumor-mediated immune suppression occurs through multiple mechanisms, including dysregulation of dendritic cell differentiation. This block in differentiation results in fewer dendritic cells and an accumulation of immunosuppressive myeloid- derived suppressor cells and is thought to contribute to tumor outgrowth and to act as an impediment to successful anti-cancer immunotherapy. Tumor-mediated myeloid dysregulation is known to be Stat3 dependent; however, the molecular mechanism of this Stat3 signaling remains poorly defined. We have previously shown that PKC betaII is required for dendritic cell differentiation. Here, we describe our finding that tumors mediate both Stat3 activation and PKC betaII down regulation in DC progenitor cells, a process mimicked by the expression of a constitutive active Stat3 mutant. This demonstrates that tumor-mediated myeloid dysregulation may be mediated by Stat3- induced PKC betaII down regulation.
Collapse
Affiliation(s)
- Matthew R Farren
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | | | | |
Collapse
|
30
|
Svajger U, Obermajer N, Jeras M. Dendritic cells treated with resveratrol during differentiation from monocytes gain substantial tolerogenic properties upon activation. Immunology 2010; 129:525-35. [PMID: 20002210 PMCID: PMC2842499 DOI: 10.1111/j.1365-2567.2009.03205.x] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2009] [Revised: 10/20/2009] [Accepted: 10/21/2009] [Indexed: 12/13/2022] Open
Abstract
Resveratrol is a polyphenol that acts on multiple molecular targets important for cell differentiation and activation. Dendritic cells (DCs) are a functionally diverse cell type and represent the most potent antigen-presenting cells of the immune system. In this study, we investigated resveratrol-induced effects on DCs during their differentiation and maturation. Our results show that resveratrol induces DC-associated tolerance, particularly when applied during DC differentiation. Costimulatory molecules CD40, CD80 and CD86 were down-regulated, as was the expression of major histocompatibility complex (MHC) class II molecules. Surface expression of inhibitory immunoglobulin-like transcript 3 (ILT3) and ILT4 molecules was induced, while human leucocyte antigen (HLA)-G expression was not affected. Resveratrol-treated DCs lost the ability to produce interleukin (IL)-12p70 after activation, but had an increased ability to produce IL-10. Such DCs were poor stimulators of allogeneic T cells and had lowered ability to induce CD4(+) T-cell migration. Furthermore, treated cells were able to generate allogeneic IL-10-secreting T cells, but were not competent in inducing FoxP3 expression These tolerogenic effects are probably associated with the effect of resveratrol on multiple molecular targets through which it interferes with DC differentiation and nuclear factor (NF)-kappaB translocation. Our data provide new insights into the molecular and functional mechanisms of the tolerogenic effects that resveratrol exerts on DCs.
Collapse
Affiliation(s)
- Urban Svajger
- Blood Transfusion Center of Slovenia, Slajmerjeva 6, Ljubljana, Slovenia.
| | | | | |
Collapse
|
31
|
Vascular endothelial growth factor stimulates protein kinase CbetaII expression in chronic lymphocytic leukemia cells. Blood 2010; 115:4447-54. [PMID: 20164467 DOI: 10.1182/blood-2009-06-229872] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Chronic lymphocytic leukemia (CLL) is a malignant disease of mature B lymphocytes. We have previously shown that a characteristic feature of CLL cells are high levels of expression and activity of protein kinase CbetaII (PKCbetaII), and that this might influence disease progression by modulating signaling in response to B-cell receptor engagement. The aim of the present work was to investigate the factors involved in stimulating PKCbetaII expression in CLL cells. Here we show that the activation of PKCbetaII in CLL cells stimulated with vascular endothelial growth factor (VEGF) can drive expression of the gene for PKCbeta, PRKCB1. We found that this effect of VEGF on PRKCB1 transcription is paralleled by high expression of PKCbetaII protein and therefore probably contributes to the malignant phenotype of CLL cells. Taken together, the data presented in this study demonstrate that VEGF, in addition to its role in providing prosurvival signals, also plays a role in overexpression of PKCbetaII, an enzyme with a specific pathophysiologic role in CLL.
Collapse
|
32
|
Kato S, Koizumi K, Yamada M, Inujima A, Takeno N, Nakanishi T, Sakurai H, Nakagawa S, Saiki I. A Phagocytotic Inducer from Herbal Constituent, Pentagalloylglucose Enhances Lipoplex-Mediated Gene Transfection in Dendritic Cells. Biol Pharm Bull 2010; 33:1878-85. [DOI: 10.1248/bpb.33.1878] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Shinichiro Kato
- Division of Pathogenic Biochemistry, Institute of Natural Medicine, University of Toyama
| | - Keiichi Koizumi
- Division of Pathogenic Biochemistry, Institute of Natural Medicine, University of Toyama
| | - Miyuki Yamada
- Division of Pathogenic Biochemistry, Institute of Natural Medicine, University of Toyama
| | - Akiko Inujima
- Division of Pathogenic Biochemistry, Institute of Natural Medicine, University of Toyama
| | - Nobuhiro Takeno
- Division of Pathogenic Biochemistry, Institute of Natural Medicine, University of Toyama
| | | | - Hiroaki Sakurai
- Division of Pathogenic Biochemistry, Institute of Natural Medicine, University of Toyama
| | - Shinsaku Nakagawa
- Department of Biotechnology and Therapeutics, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Ikuo Saiki
- Division of Pathogenic Biochemistry, Institute of Natural Medicine, University of Toyama
| |
Collapse
|
33
|
Voris JP, Sitailo LA, Rahn HR, Defnet A, Gerds AT, Sprague R, Yadav V, Caroline Le Poole I, Denning MF. Functional alterations in protein kinase C beta II expression in melanoma. Pigment Cell Melanoma Res 2009; 23:216-24. [PMID: 20015121 DOI: 10.1111/j.1755-148x.2009.00664.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Protein kinase C (PKC) is a heterogeneous family of serine/threonine protein kinases that have different biological effects in normal and neoplastic melanocytes (MCs). To explore the mechanism behind their differential response to PKC activation, we analyzed the expression profile of all nine PKC isoforms in normal human MCs, HPV16 E6/E7 immortalized MCs, and a panel of melanoma cell lines. We found reduced PKCbeta and increased PKCzeta and PKCiota expression at both the protein and mRNA levels in immortalized MCs and melanoma lines. We focused on PKCbeta as it has been functionally linked to melanin production and oxidative stress response. Re-expression of PKCbeta in melanoma cells inhibited colony formation in soft agar, indicating that PKCbeta loss in melanoma is important for melanoma growth. PKCbetaII, but not PKCbetaI, was localized to the mitochondria, and inhibition of PKCbeta significantly reduced UV-induced reactive oxygen species (ROS) in MCs with high PKCbeta expression. Thus alterations in PKCbeta expression in melanoma contribute to their neoplastic phenotype, possibly by reducing oxidative stress, and may constitute a selective therapeutic target.
Collapse
Affiliation(s)
- John P Voris
- The Oncology Institute, Loyola University Chicago, Maywood, IL, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Lintas C, Sacco R, Garbett K, Mirnics K, Militerni R, Bravaccio C, Curatolo P, Manzi B, Schneider C, Melmed R, Elia M, Pascucci T, Puglisi-Allegra S, Reichelt KL, Persico AM. Involvement of the PRKCB1 gene in autistic disorder: significant genetic association and reduced neocortical gene expression. Mol Psychiatry 2009; 14:705-18. [PMID: 18317465 DOI: 10.1038/mp.2008.21] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Protein kinase C enzymes play an important role in signal transduction, regulation of gene expression and control of cell division and differentiation. The fsI and betaII isoenzymes result from the alternative splicing of the PKCbeta gene (PRKCB1), previously found to be associated with autism. We performed a family-based association study in 229 simplex and 5 multiplex families, and a postmortem study of PRKCB1 gene expression in temporocortical gray matter (BA41/42) of 11 autistic patients and controls. PRKCB1 gene haplotypes are significantly associated with autism (P<0.05) and have the autistic endophenotype of enhanced oligopeptiduria (P<0.05). Temporocortical PRKCB1 gene expression was reduced on average by 35 and 31% for the PRKCB1-1 and PRKCB1-2 isoforms (P<0.01 and <0.05, respectively) according to qPCR. Protein amounts measured for the PKCbetaII isoform were similarly decreased by 35% (P=0.05). Decreased gene expression characterized patients carrying the 'normal' PRKCB1 alleles, whereas patients homozygous for the autism-associated alleles displayed mRNA levels comparable to those of controls. Whole genome expression analysis unveiled a partial disruption in the coordinated expression of PKCbeta-driven genes, including several cytokines. These results confirm the association between autism and PRKCB1 gene variants, point toward PKCbeta roles in altered epithelial permeability, demonstrate a significant downregulation of brain PRKCB1 gene expression in autism and suggest that it could represent a compensatory adjustment aimed at limiting an ongoing dysreactive immune process. Altogether, these data underscore potential PKCbeta roles in autism pathogenesis and spur interest in the identification and functional characterization of PRKCB1 gene variants conferring autism vulnerability.
Collapse
Affiliation(s)
- C Lintas
- Laboratory of Molecular Psychiatry and Neurogenetics, University Campus Bio-Medico, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Kelly DJ, Edgley AJ, Zhang Y, Thai K, Tan SM, Cox AJ, Advani A, Connelly KA, Whiteside CI, Gilbert RE. Protein kinase C-beta inhibition attenuates the progression of nephropathy in non-diabetic kidney disease. Nephrol Dial Transplant 2009; 24:1782-90. [PMID: 19155535 DOI: 10.1093/ndt/gfn729] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Activation of protein kinase C (PKC) has been implicated in the pathogenesis of diabetic nephropathy where therapy targeting the beta isoform of this enzyme is in advanced clinical development. However, PKC-beta is also increased in various forms of human glomerulonephritis with several potentially nephrotoxic factors, other than high glucose, resulting in PKC-beta activation. Accordingly, we sought to examine the effects of PKC-beta inhibition in a non-diabetic model of progressive kidney disease. METHODS Subtotally nephrectomized (STNx) rats were randomly assigned to receive either the selective PKC-beta inhibitor, ruboxistaurin or vehicle. In addition to functional and structural parameters, gene expression of the podocyte slit-pore diaphragm protein, nephrin, was also assessed. RESULTS STNx animals developed hypertension, proteinuria and reduced glomerular filtration rate (GFR) in association with marked glomerulosclerosis and tubulointerstitial fibrosis. Glomerular nephrin expression was also reduced. Without affecting blood pressure, ruboxistaurin treatment attenuated the impairment in GFR and reduced the extent of both glomerulosclerosis and tubulointerstitial fibrosis in STNx rats. In contrast, neither proteinuria nor the reduction in nephrin expression was improved by ruboxistaurin. CONCLUSIONS These findings indicate firstly that PKC-beta inhibition may provide a new therapeutic strategy in non-diabetic kidney disease and secondly that improvement in GFR is not inextricably linked to reduction in proteinuria.
Collapse
Affiliation(s)
- Darren J Kelly
- Department of Medicine, University of Melbourne, St Vincent's Hospital, Fitzroy, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Chen C, Johnston TD, Jeon H, Gedaly R, McHugh P, Ranjan D. Cyclosporin A up-regulates and activates protein kinase C-zeta in EBV-infected and EBV-transformed human B-cells. J Surg Res 2008; 153:156-61. [PMID: 18486150 DOI: 10.1016/j.jss.2008.03.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2008] [Revised: 02/12/2008] [Accepted: 03/05/2008] [Indexed: 10/22/2022]
Abstract
BACKGROUND Protein Kinase C (PKC) is a family of enzymes that plays a key role in cell signaling pathways leading to cellular activation and proliferation. Conventional PKC (cPKC) is dependent on calcium for activation. We have proposed that cyclosporin A (CsA), despite being a calcineurin inhibitor, will activate PKC in B cells, thus promoting Epstein-Barr virus (EBV)-induced transformation. Here we show that CsA promoted atypical PKC isoform PKC-zeta in B cells. MATERIALS AND METHODS Western-blot was used to assay PKC-zeta protein level in EBV-B cells. Confocal microscopy was used to assay PKC-zeta translocation from cytosol to cell membrane, a known process of PKC activation. RESULTS CsA (500 ng/mL) time dependently increased PKC-zeta from control of 7055 units to 7145, 10,805, 10,914, and 12,705 units, respectively, after 15 min, 1 h, 12 h, and 24 h of incubation in EBV-transformed human B-cell line (LCL). CsA increased PKC-zeta expression was inhibited 50% by Vit.E (40 microM) indicating that this effect may be due to oxidative stress induced by CsA. Indeed, after oxidant H(2)O(2) (0.1 mM) treatment, PKC-zeta protein level in LCL cells increased 124%, 257%, 349%, and 359% after 15 min, 1 h, 12 h, and 24 h of culture compared with control. Addition of Vit.E (40 microM) in H(2)O(2) (0.1 mM) treatment and then with Vit.E in the culture decreased PKC-zeta level in LCL cells 26%, 20%, 41%, and 60% after 15 min, 1 h, 12 h, and 24 h of culture. In confocal microscopy in Jurkat T cell line, phorbol 12-myristate 13-acetate (PMA) activated cPKC isoform PKCalpha after 30 min treatment and activated PKC-zeta after 60 min treatment. CsA inhibited PMA activation of PKC-alpha, but not PKC-zeta. CsA alone did not activate PKC-alpha or PKC-zeta in Jurkat T cells. In LCL and in EBV-infected human B-cells, PMA stimulated PKC-alpha activation after 30 min treatment and stimulated PKC-zeta activation after 60 min treatment. CsA inhibited PMA activation of PKC-alpha, but not PKC-zeta. In addition, CsA activated PKC-zeta in the EBV-transformed and EBV-infected human B cells. CONCLUSION These experiments show that CsA-induced oxidative stress caused PKC-zeta up-regulation in LCL cells, and show the differential effect of CsA in the PKC signaling pathways in T cells versus B cells. CsA-induced PKC-zeta activation may be an important signaling step in EBV-induced post-transplant lymphoproliferative disorders.
Collapse
Affiliation(s)
- Changguo Chen
- Department of Surgery, University of Kentucky, College of Medicine, Lexington, KY 40536, USA
| | | | | | | | | | | |
Collapse
|
37
|
Lindner I, Cejas PJ, Carlson LM, Torruellas J, Plano GV, Lee KP. Signal transduction in DC differentiation: winged messengers and Achilles' heel. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 590:1-29. [PMID: 17191374 DOI: 10.1007/978-0-387-34814-8_1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Inna Lindner
- University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | | | | | | | | | | |
Collapse
|
38
|
Wakui M, Nakano K, Matsushita S. Notch ligand mRNA levels of human APCs predict Th1/Th2-promoting activities. Biochem Biophys Res Commun 2007; 358:596-601. [PMID: 17499611 DOI: 10.1016/j.bbrc.2007.04.175] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2007] [Accepted: 04/27/2007] [Indexed: 11/18/2022]
Abstract
Although the contribution of Notch ligands expressed by antigen-presenting cells (APCs) to the Th1/Th2 differentiation has been suggested in mouse studies, their nature in humans remains obscure. To assess the issue, we examined correlation of Notch ligand mRNA levels of human APCs with Th1/Th2-promoting stimulations, i.e. Th1/Th2 adjuvant activities. The expression patterns of human dendritic cells (DCs) and leukemic cell line models of APCs differed partly from those previously observed in mouse DCs and by cellular types and maturation stages. Most strikingly, Th2 adjuvants enhanced the Delta1 expression on human APCs but did not induce Delta4 expression, which was induced by a Th1 adjuvant. The differential expression patterns suggest a distinct role of these Delta members in Th1/Th2 differentiation and their predictive potential for Th1/Th2-promoting activities.
Collapse
Affiliation(s)
- Masatoshi Wakui
- Department of Allergy and Immunology, Faculty of Medicine, Saitama Medical University, 38 Morohongo, Moroyama, Saitama, Japan
| | | | | |
Collapse
|
39
|
Lin YF, Lee HM, Leu SJ, Tsai YH. The essentiality of PKCα and PKCβI translocation for CD14+monocyte differentiation towards macrophages and dendritic cells, respectively. J Cell Biochem 2007; 102:429-41. [PMID: 17455194 DOI: 10.1002/jcb.21305] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Human peripheral CD14(+)monocytes have been known to differentiate into monocyte-derived macrophages (MDMs) or dendritic cells (MoDCs) upon suitable stimulation. However, the key intracellular molecule(s) associated with their differentiation towards specific cell types was(were) not fully understood. This study was designated to determine the association of PKC isoenzymes with the differentiation of CD14(+)monocytes into MDMs or MoDCs. Purified human peripheral CD14(+)monocytes were cultured with GM-CSF, or GM-CSF plus IL-4 for 7 days to induce cell differentiation. The phenotypic changes were analyzed by Flow-Cytometry using various specific antibodies to cell type-specific surface markers. The immunological functions of these differentiated cells were determined by measuring the amounts of TNF-alpha secretion for MDMs, and the capacities of antigen-capturing and bacterial phagocytosis for MoDCs. The translocations of PKC isoenzymes in these cells from cytosol to plasma membrane were examined by Western Blot analysis and Confocal Microscopic observation. The treatment of CD14(+)monocytes with either GM-CSF or PMA elicited PKCalpha translocation and consequently induced their differentiation into MDMs. The inclusion of PKCalpha/beta(I) specific inhibitor, Go6976, greatly inhibited the GM-CSF-induced PKCalpha translocation and dose-dependently reduced the GM-CSF- induced MDM differentiation. On the other hand, the simultaneous pretreatment of CD14(+)monocytes with Go6976 and PKCbeta-specific inhibitor predominantly suppressed the GM-CSF/IL-4-induced generation of MoDCs. Further study demonstrated that GM-CSF/IL-4 selectively induced the translocation of PKCbeta(I), not PKCalpha or PKCbeta(II), in CD14(+)monocytes. In conclusion, the cell fate commitment of CD14(+)monocytes towards MDMs or MoDCs appears to be steered by the selective activation of PKCalpha or PKCbeta(I), respectively.
Collapse
Affiliation(s)
- Yuan-Feng Lin
- Graduate Institute of Pharmaceutical Science, Taipei Medical University, Taipei, Taiwan
| | | | | | | |
Collapse
|
40
|
Tomic J, White D, Shi Y, Mena J, Hammond C, He L, Miller RL, Spaner DE. Sensitization of IL-2 Signaling through TLR-7 Enhances B Lymphoma Cell Immunogenicity. THE JOURNAL OF IMMUNOLOGY 2006; 176:3830-9. [PMID: 16517754 DOI: 10.4049/jimmunol.176.6.3830] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The innate ability of B lymphoma cells to escape control by tumor-reactive T cells must be overcome to develop effective immunotherapies for these diseases. Because signals from both the innate and adaptive immune systems direct the acquisition of strong immunogenicity by professional APCs, the effects of IL-2 and the TLR-7 agonist, S28690, on the immunogenic properties of chronic lymphocytic leukemia (CLL) B cells were studied. IL-2 with S28690 caused CLL cells to proliferate and increased their expression of B7-family members, production of TNF-alpha and IL-10, and levels of tyrosine-phosphorylated STAT-1 and STAT-3 proteins. S28690 increased CD25 expression on CLL cells and sensitized them to IL-2 signaling. However, IL-2 did not change TLR-7 expression or signaling in CLL cells. The ability to stimulate T cell proliferation required additional activation of protein kinase C, which inhibited tumor cell proliferation, "switched off" IL-10 production, and caused essentially all CLL cells (regardless of clinical stage) to acquire a CD83(high)CD80(high)CD86(high)CD54(high) surface phenotype marked by the activation of STAT-1 without STAT-3. These findings suggest that TLR-7 "licenses" human B cells to respond to cytokines of the adaptive immune system (such as IL-2) and provide a strategy to increase the immunogenicity of lymphoma cells for therapeutic purposes.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Cells, Cultured
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Imidazoles/pharmacology
- Interleukin-2/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Lymphoma, B-Cell/immunology
- Lymphoma, B-Cell/metabolism
- Lymphoma, B-Cell/pathology
- Male
- Middle Aged
- Phenotype
- Protein Kinase C/metabolism
- Quinolines/pharmacology
- Receptors, Interleukin-2/genetics
- STAT1 Transcription Factor/metabolism
- STAT3 Transcription Factor/metabolism
- Signal Transduction
- Toll-Like Receptor 7/immunology
- Toll-Like Receptor 7/metabolism
Collapse
Affiliation(s)
- Jelena Tomic
- Division of Molecular and Cellular Biology, Research Institute, Sunnybrook and Women's College Health Sciences Center, 2075 Bayview Avenue, Toronto, Ontario M4N 3M5, Canada
| | | | | | | | | | | | | | | |
Collapse
|