1
|
Lin J, Chen X, Li Y, Yu L, Chen Y, Zhang B. A dual-targeting therapeutic nanobubble for imaging-guided atherosclerosis treatment. Mater Today Bio 2024; 26:101037. [PMID: 38586870 PMCID: PMC10995877 DOI: 10.1016/j.mtbio.2024.101037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/05/2024] [Accepted: 03/19/2024] [Indexed: 04/09/2024] Open
Abstract
Atherosclerosis is a cardiovascular disease that seriously endangers human health. Low shear stress (LSS) is recognized as a vital factor in causing chronic inflammatory and further inducing the occurrence and development of atherosclerosis. Targeting imaging and treatment are of substantial significance for the diagnosis and therapy of atherosclerosis. On this ground, a kind of ultrasound (US) imaging-guided therapeutic polymer nanobubbles (NBs) with dual targeting of magnetism and antibody was rationally designed and constructed for the efficiently treating LSS-mediated atherosclerosis. Under the combined targeting effect of an external magnetic field and antibodies, the drug-loaded therapeutic NBs can be effectively accumulated in the inflammatory area caused by LSS. Upon US irradiation, the NBs can be selectively disrupted, leading to the rapid release of the loaded drugs at the targeted site. Notably, the US irradiation generates a cavitation effect that induces repairable micro gaps in nearby cells, thereby enhancing the uptake of released drugs and further improving the therapeutic effect. The prominent US imaging, efficient anti-inflammatory effect and treatment outcome of LSS-mediated atherosclerosis had been verified in vivo on a surgically constructed LSS-atherosclerosis animal model. This work showcased the potential of the designed NBs with multifunctionality for in vivo imaging, dual-targeting, and drug delivery in the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Jie Lin
- Department of Ultrasound, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, PR China
| | - Xiaoying Chen
- Department of Ultrasound, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, PR China
| | - Yi Li
- Department of Ultrasound, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, PR China
| | - Luodan Yu
- Department of Radiology, Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, PR China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China
- Shanghai Institute of Materdicine, Shanghai, 200051, PR China
| | - Bo Zhang
- Department of Ultrasound, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, PR China
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, PR China
| |
Collapse
|
2
|
Zhang Y, Zhou H, Chen X, Wang N, Zhan Y, Huang Z, Ruan K, Qi Q, Deng M, Jiang Y. A novel tRNA-derived fragment tRF-3023b suppresses inflammation in RAW264.7 cells by targeting Cul4a through NF-κB signaling. Funct Integr Genomics 2024; 24:9. [PMID: 38221594 DOI: 10.1007/s10142-024-01285-3] [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: 09/28/2023] [Revised: 11/29/2023] [Accepted: 01/01/2024] [Indexed: 01/16/2024]
Abstract
The role of transfer RNA (tRNA)-derived fragment (tRF) in various diseases has been established. However, the effect of tRF-3023b on inflammation remains unclear. Inflammation was imitated in RAW264.7 cells by adding Lipopolysaccharide (LPS). Cells were first divided into control, LPS, and LPS + Bulleyaconitine A (BLA) groups. The contents of TNF-α, IL-6, and MCP-1 were quantified using ELISA. The levels of cyclooxygenase-2 (COX2), inducible nitric oxide synthase (iNOS), and the phosphorylation of nuclear factor-kappa B (NF-κB)-P65 (p-P65) were detected by Western blotting. RNA sequencing was utilized to find differentially expressed tRFs (DE-tRFs) among three groups. The levels of various tRFs were checked by quantitative real-time PCR (qRT-PCR). Cell cycle and apoptosis were checked by flow cytometry. Dluciferase reporter assay was applied to predict and confirm the interaction between tRF-3023b and Cullin 4A (Cul4a), subsequently RNA pull-down followed by mass spectrometry analysis were conducted. BLA treatment decreased the contents of TNF-α, IL-6, MCP-1, and the expression levels of COX2, iNOS, p-P65. We found 6 DE-tRFs in LPS + BLA group compared to LPS group, tRF-3023b was high expression in control and BLA groups, and the lowest in LPS group. Cul4a was a direct target of tRF-3023b. tRF-3023b mimic affected the cell cycle distribution, promoted cells apoptosis, and suppressed the TNF-α, IL-6, MCP-1, COX2, iNOS and p-P65. The suppression of Cul4a affected the cell cycle distribution, resulted in an increase of cell apoptosis while a decrease of TNF-α, IL-6, MCP-1, COX2, iNOS and p-P65. Furthermore, Cul4a overexpression reversed the effect of tRF-3023b mimic. Cul4a knockdown reversed the effect of tRF-3023b inhibitor. Our study positions tRF-3023b as a compelling candidate, through its interaction with Cul4a, the underlying mechanism on inflammation maybe related to NF-κB pathway. The study provides a basis for exploring new therapeutic strategies for inflammation.
Collapse
Affiliation(s)
- Ying Zhang
- Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University, No. 118 Jiahang Road, Nanhu District, Jiaxing, 314001, China
| | - Hua Zhou
- Department of Physiology, Anhui Medical College, Hefei, China
| | - Xu Chen
- Department of Pathogen Biology and Immunology, Jiaxing University School of Medicine, No. 118 Jiahang Road, Nanhu District, Jiaxing, 314001, China
| | - Ningning Wang
- Department of Pathogen Biology and Immunology, Jiaxing University School of Medicine, No. 118 Jiahang Road, Nanhu District, Jiaxing, 314001, China
| | - Yunfei Zhan
- Department of Pathogen Biology and Immunology, Jiaxing University School of Medicine, No. 118 Jiahang Road, Nanhu District, Jiaxing, 314001, China
| | - Ziyi Huang
- Department of Pathogen Biology and Immunology, Jiaxing University School of Medicine, No. 118 Jiahang Road, Nanhu District, Jiaxing, 314001, China
| | - Kaiyi Ruan
- Department of Pathogen Biology and Immunology, Jiaxing University School of Medicine, No. 118 Jiahang Road, Nanhu District, Jiaxing, 314001, China
| | - Qiulan Qi
- Department of Pathogen Biology and Immunology, Jiaxing University School of Medicine, No. 118 Jiahang Road, Nanhu District, Jiaxing, 314001, China.
| | - Min Deng
- Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University, No. 118 Jiahang Road, Nanhu District, Jiaxing, 314001, China.
- Department of Infectious Diseases, Affiliated Hospital of Jiaxing University, No. 1882 South Zhonghuan Road, Nanhu District, Jiaxing, 314000, China.
| | - Yuxin Jiang
- Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University, No. 118 Jiahang Road, Nanhu District, Jiaxing, 314001, China.
- Department of Pathogen Biology and Immunology, Jiaxing University School of Medicine, No. 118 Jiahang Road, Nanhu District, Jiaxing, 314001, China.
| |
Collapse
|
3
|
The critical role of T cells in glucocorticoid-induced osteoporosis. Cell Death Dis 2020; 12:45. [PMID: 33414409 PMCID: PMC7791068 DOI: 10.1038/s41419-020-03249-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 12/14/2022]
Abstract
Glucocorticoids (GC) are widely used clinically, despite the presence of significant side effects, including glucocorticoid-induced osteoporosis (GIOP). While GC are believed to act directly on osteoblasts and osteoclasts to promote osteoporosis, the detailed underlying molecular mechanism of GC-induced osteoporosis is still not fully elucidated. Here, we show that lymphocytes play a pivotal role in regulating GC-induced osteoporosis. We show that GIOP could not be induced in SCID mice that lack T cells, but it could be re-established by adoptive transfer of splenic T cells from wild-type mice. As expected, T cells in the periphery are greatly reduced by GC; instead, they accumulate in the bone marrow where they are protected from GC-induced apoptosis. These bone marrow T cells in GC-treated mice express high steady-state levels of NF-κB receptor activator ligand (RANKL), which promotes the formation and maturation of osteoclasts and induces osteoporosis. Taken together, these findings reveal a critical role for T cells in GIOP.
Collapse
|
4
|
Mosser DM, Hamidzadeh K, Goncalves R. Macrophages and the maintenance of homeostasis. Cell Mol Immunol 2020; 18:579-587. [PMID: 32934339 PMCID: PMC7491045 DOI: 10.1038/s41423-020-00541-3] [Citation(s) in RCA: 280] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 08/17/2020] [Indexed: 12/17/2022] Open
Abstract
There have been many chapters written about macrophage polarization. These chapters generally focus on the role of macrophages in orchestrating immune responses by highlighting the T-cell-derived cytokines that shape these polarizing responses. This bias toward immunity is understandable, given the importance of macrophages to host defense. However, macrophages are ubiquitous and are involved in many different cellular processes, and describing them as immune cells is undoubtedly an oversimplification. It disregards their important roles in development, tissue remodeling, wound healing, angiogenesis, and metabolism, to name just a few processes. In this chapter, we propose that macrophages function as transducers in the body. According to Wikipedia, “A transducer is a device that converts energy from one form to another.” The word transducer is a term used to describe both the “sensor,” which can interpret a wide range of energy forms, and the “actuator,” which can switch voltages or currents to affect the environment. Macrophages are able to sense a seemingly endless variety of inputs from their environment and transduce these inputs into a variety of different response outcomes. Thus, rather than functioning as immune cells, they should be considered more broadly as cellular transducers that interpret microenvironmental changes and actuate vital tissue responses. In this chapter, we will describe some of the sensory stimuli that macrophages perceive and the responses they make to these stimuli to achieve their prime directive, which is the maintenance of homeostasis.
Collapse
Affiliation(s)
- David M Mosser
- The Department of Cell Biology and Molecular Genetics, The University of Maryland, College Park, MD, 20742, USA.
| | - Kajal Hamidzadeh
- The Department of Cell Biology and Molecular Genetics, The University of Maryland, College Park, MD, 20742, USA
| | - Ricardo Goncalves
- The Department of General Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| |
Collapse
|
5
|
Chawla AS, Khalsa JK, Dhar A, Gupta S, Umar D, Arimbasseri GA, Bal V, George A, Rath S. A role for cell-autocrine interleukin-2 in regulatory T-cell homeostasis. Immunology 2020; 160:295-309. [PMID: 32187647 DOI: 10.1111/imm.13194] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 03/16/2020] [Accepted: 03/16/2020] [Indexed: 12/30/2022] Open
Abstract
Activated T-cells make both interleukin-2 (IL2) and its high-affinity receptor component CD25. Regulatory CD4 T-cells (Treg cells) do not make IL2, and the IL2-CD25 circuit is considered a paracrine circuit crucial in their generation and maintenance. Yet, all T-cells are capable of making IL2 at some stage during differentiation, making a cell-intrinsic autocrine circuit additionally possible. When we re-visited experiments with mixed bone marrow chimeras using a wide range of ratios of wild-type (WT) and IL2-/- genotype progenitors, we found that, as expected, thymic Treg cells were almost equivalent between WT and IL2-/- genotypes at ratios with WT prominence. However, at WT-limiting ratios, the IL2-/- genotype showed lower thymic Treg frequencies, indicating a role for cell-intrinsic autocrine IL2 in thymic Treg generation under IL2-limiting conditions. Further, peripheral IL2-/- naive CD4 T-cells showed poor conversion to inducible Tregs (pTregs) both in vivo and in vitro, again indicating a significant role for cell-intrinsic autocrine IL2 in their generation. Peripherally, the IL2-/- genotype was less prominent at all WT:IL2-/- ratios among both thymic Tregs (tTregs) and pTregs, adoptively transferred IL2-/- Tregs showed poorer survival than WT Tregs did, and RNA-seq analysis of WT and IL2-/- Tregs showed interesting differences in the T-cell receptor and transforming growth factor-beta-bone morphogenetic protein-JNK pathways between them, suggesting a non-titrating role for cell-intrinsic autocrine IL2 in Treg programming. These data indicate that cell-intrinsic autocrine IL2 plays significant roles in Treg generation and maintenance.
Collapse
Affiliation(s)
| | | | - Atika Dhar
- National Institute of Immunology, New Delhi, India
| | - Suman Gupta
- National Institute of Immunology, New Delhi, India
| | - Danish Umar
- National Institute of Immunology, New Delhi, India
| | | | - Vineeta Bal
- National Institute of Immunology, New Delhi, India
| | - Anna George
- National Institute of Immunology, New Delhi, India
| | | |
Collapse
|
6
|
Goto K, Hiramoto K, Ooi K. Th2 and Th17 Induce Dry Skin in a Mouse Model of Arthritis. Biol Pharm Bull 2019; 42:468-474. [PMID: 30828078 DOI: 10.1248/bpb.b18-00803] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Skin dryness is a characteristic of rheumatoid arthritis (RA) model mice. However, the mechanism underlying the induction of dry skin by RA is unclear. We hypothesized that T helper (Th)2 and Th17 cells mediate this process. A mouse model of DBA/1JJmsSlc collagen-induced arthritis was treated with Th2 or Th17 cell inhibitor, and transepidermal water loss (TEWL) and the expression of markers associated with allergic reaction and inflammation were evaluated. TEWL and plasma levels of thymic stromal lymphopoietin, interleukin (IL)-6 and -17, and tumor necrosis factor (TNF)-α were increased in the arthritis mouse model compared to that in control mice. Administration of Th2 cell inhibitor abolished the increase in TEWL, IL-6, and TNF-α levels, whereas Th17 cell inhibitor reversed TEWL and decreased IL-17 level. Th2 and Th17 cells contribute to the induction of dry skin, but via distinct mechanisms.
Collapse
Affiliation(s)
- Kenji Goto
- Laboratory of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science.,Laboratory of Pathophysiology and Pharmacotherapy, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science
| | - Keiichi Hiramoto
- Laboratory of Pathophysiology and Pharmacotherapy, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science
| | - Kazuya Ooi
- Laboratory of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science.,Laboratory of Pathophysiology and Pharmacotherapy, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science
| |
Collapse
|
7
|
Amôr NG, de Oliveira CE, Gasparoto TH, Vilas Boas VG, Perri G, Kaneno R, Lara VS, Garlet GP, da Silva JS, Martins GA, Hogaboam C, Cavassani KA, Campanelli AP. ST2/IL-33 signaling promotes malignant development of experimental squamous cell carcinoma by decreasing NK cells cytotoxicity and modulating the intratumoral cell infiltrate. Oncotarget 2018; 9:30894-30904. [PMID: 30112116 PMCID: PMC6089399 DOI: 10.18632/oncotarget.25768] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 05/19/2018] [Indexed: 02/06/2023] Open
Abstract
Squamous cell carcinoma (SCC) is the second most common form of skin cancer and the mechanism(s) involved in the progression of this tumor are unknown. Increases in the expression of IL-33/ST2 axis components have been demonstrated to contribute to neoplastic transformation in several tumor models and interleukin-33 is correlated with poor prognosis of patients with squamous cell carcinoma of the tongue. Based on these observations, we sought to determine the role of the IL-33/ST2 pathway during the development of SCC. Our findings show that ST2-deficiency led to a marked decrease in the severity of skin lesions, suggesting that ST2 signaling contributed to tumor development. An analysis of tumor lesions in wild-type and ST2KO mice revealed that a lack of ST2 was associated with specific and significant reductions in the numbers of CD4+ T cells, CD8+ T cells, dendritic cells, and macrophages. In addition, NK cells that were isolated from ST2KO mice exhibited higher cytotoxic activity than cells isolated from wild-type mice. Notably, ST2 deficiency resulted in lower IFN-γ, TNF-α, IL-10, and IL-17 production in tumor samples. Our findings indicate that the IL-33/ST2 pathway contributes to the development of SCC by affecting leukocyte migration to tumor microenvironment and impairing NK cytotoxic activity.
Collapse
Affiliation(s)
- Nádia Ghinelli Amôr
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Dr. Octávio Pinheiro Brisolla, Bauru, SP, 17012-901, Brazil
| | - Carine Ervolino de Oliveira
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Dr. Octávio Pinheiro Brisolla, Bauru, SP, 17012-901, Brazil
| | - Thaís Helena Gasparoto
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Dr. Octávio Pinheiro Brisolla, Bauru, SP, 17012-901, Brazil
| | - Vanessa Garcia Vilas Boas
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Dr. Octávio Pinheiro Brisolla, Bauru, SP, 17012-901, Brazil
| | - Graziela Perri
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Dr. Octávio Pinheiro Brisolla, Bauru, SP, 17012-901, Brazil
| | - Ramon Kaneno
- Department of Microbiology and Immunology, Institute of Biosciences of Botucatu, São Paulo State University, R. Prof. Dr. Antônio Celso Wagner Zanin, Botucatu, SP, 18618-689, Brazil
| | - Vanessa Soares Lara
- Department of Stomatology - Oral Pathology, Bauru School of Dentistry, University of São Paulo, Al. Dr. Octávio Pinheiro Brisolla, Bauru, SP, 17012-901, Brazil
| | - Gustavo Pompermaier Garlet
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Dr. Octávio Pinheiro Brisolla, Bauru, SP, 17012-901, Brazil
| | - João Santana da Silva
- Department of Biochemistry and Immunology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Gislâine A Martins
- Department of Biomedical Sciences (Research Division of Immunology) and Medicine, F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Cory Hogaboam
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Karen A Cavassani
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Ana Paula Campanelli
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Dr. Octávio Pinheiro Brisolla, Bauru, SP, 17012-901, Brazil
| |
Collapse
|
8
|
Huang C, Li W, Zhang Q, Chen L, Chen W, Zhang H, Ni Y. Anti-inflammatory activities of Guang-Pheretima extract in lipopolysaccharide-stimulated RAW 264.7 murine macrophages. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 18:46. [PMID: 29391009 PMCID: PMC5795835 DOI: 10.1186/s12906-018-2086-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 01/14/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Guang-Pheretima, which is originated from Pheretima aspergillum, has been documented in academic Chinese herbal studies for nearly 2000 years for its prominent treating effects of various inflammatory diseases such as asthma, cough and fever. However, the anti-inflammatory activity and mechanism of Guang-Pheretima has been rarely reported. Hence, we investigated the inhibitory effect and the underlying mechanism of Guang-Pheretima aqueous extracts on inflammatory response in RAW 264.7 cells. METHOD RAW 264.7 macrophages were pretreated with various concentrations of Guang-Pheretima decoction (GPD) or protein-free Guang-Pheretima decoction (PF-GPD) and subsequently stimulated with lipopolysaccharide (LPS) to trigger the inflammatory response. Productions of nitric oxide (NO) were determined by Griess reaction, and prostaglandin E2 (PGE2), tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, IL-6 were measured by enzyme-linked immunosorbent assays (ELISA). The protein expressions and messenger ribonucleic acid (mRNA) amounts of inducible nitric oxide synthase (iNOS) and cyclooxygenase (COX)-2 were analyzed by Western Blot and Real-Time polymerase chain reaction (PCR), respectively. Finally, the translocation of nuclear factor (NF)-κB was observed by Western Blot. RESULTS GPD of the experimental concentrations showed no anti-inflammatory activity. In contrast, PF-GPD at concentrations of 40-320 μg/mL significantly inhibited NF-κB activation and reduced the production of inflammatory mediators, such as NO, PGE2, TNF-α, as well as the related key synthases including iNOS and COX-2. Moreover, PF-GPD markedly suppressed the release of inflammatory cytokines, such as IL-1β and IL-6. CONCLUSION These results demonstrate the excellent anti-inflammatory properties of PF-GPD, and suggest that Guang-Pheretima may be used to treat and prevent certain inflammatory diseases.
Collapse
Affiliation(s)
- Chuanqi Huang
- Department of Pharmacy, Wuhan No.1 Hospital (Wuhan Integrated TCM & Western Medicine Hospital), 215 Zhongshan Avenue, Wuhan, 430022, China
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou Higher Education Mega Center, 232 Wai Huan Road East, Guangzhou, 510006, China
| | - Wei Li
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou Higher Education Mega Center, 232 Wai Huan Road East, Guangzhou, 510006, China.
| | - Qiufeng Zhang
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou Higher Education Mega Center, 232 Wai Huan Road East, Guangzhou, 510006, China
| | - Lihong Chen
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou Higher Education Mega Center, 232 Wai Huan Road East, Guangzhou, 510006, China
| | - Weiming Chen
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou Higher Education Mega Center, 232 Wai Huan Road East, Guangzhou, 510006, China
| | - Hongchao Zhang
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou Higher Education Mega Center, 232 Wai Huan Road East, Guangzhou, 510006, China
| | - Yuxin Ni
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou Higher Education Mega Center, 232 Wai Huan Road East, Guangzhou, 510006, China
| |
Collapse
|
9
|
Ginaldi L, De Martinis M. Osteoimmunology and Beyond. Curr Med Chem 2017; 23:3754-3774. [PMID: 27604089 PMCID: PMC5204071 DOI: 10.2174/0929867323666160907162546] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 09/02/2016] [Accepted: 09/06/2016] [Indexed: 12/27/2022]
Abstract
Abstract: Objective Osteoimmunology investigates interactions between skeleton and immune system. In the light of recent discoveries in this field, a new reading register of osteoporosis is actually emerging, in which bone and immune cells are strictly interconnected. Osteoporosis could therefore be considered a chronic immune mediated disease which shares with other age related disorders a common inflammatory background. Here, we highlight these recent discoveries and the new landscape that is emerging. Method Extensive literature search in PubMed central. Results While the inflammatory nature of osteoporosis has been clearly recognized, other interesting aspects of osteoimmunology are currently emerging. In addition, mounting evidence indicates that the immunoskeletal interface is involved in the regulation of important body functions beyond bone remodeling. Bone cells take part with cells of the immune system in various immunological functions, configuring a real expanded immune system, and are therefore variously involved not only as target but also as main actors in various pathological conditions affecting primarily the immune system, such as autoimmunity and immune deficiencies, as well as in aging, menopause and other diseases sharing an inflammatory background. Conclusion The review highlights the complexity of interwoven pathways and shared mechanisms of the crosstalk between the immune and bone systems. More interestingly, the interdisciplinary field of osteoimmunology is now expanding beyond bone and immune cells, defining new homeostatic networks in which other organs and systems are functionally interconnected. Therefore, the correct skeletal integrity maintenance may be also relevant to other functions outside its involvement in bone mineral homeostasis, hemopoiesis and immunity.
Collapse
Affiliation(s)
- Lia Ginaldi
- School and Unit of Allergy and Clinical Immunology, Department of Life, Health, & Environmental Sciences, University of L'Aquila, Italy.
| | | |
Collapse
|
10
|
İlhan E, Kaplan A, Güvenç TS, Biteker M, Karabulut E, Işıklı S. Electrocardiographic features of patients with earthquake related posttraumatic stress disorder. World J Cardiol 2013; 5:60-64. [PMID: 23538549 PMCID: PMC3610008 DOI: 10.4330/wjc.v5.i3.60] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 10/25/2012] [Accepted: 01/21/2013] [Indexed: 02/06/2023] Open
Abstract
AIM: To analyze electrocardiographic features of patients diagnosed with posttraumatic stress disorder (PTSD) after the Van-Erciş earthquake, with a shock measuring 7.2 on the Richter scale that took place in Turkey in October 2011.
METHODS: Surface electrocardiograms of 12 patients with PTSD admitted to Van Erciş State Hospital (Van, Turkey) from February 2012 to May 2012 were examined. Psychiatric interviews of the sex and age matched control subjects, who had experienced the earthquake, confirmed the absence of any known diagnosable psychiatric conditions in the control group.
RESULTS: A wide range of electrocardiogram (ECG) parameters, such as P-wave dispersion, QT dispersion, QT interval, Tpeak to Tend interval, intrinsicoid deflection durations and other traditional parameters were similar in both groups. There was no one with an abnormal P wave axis, short or long PR interval, long or short QT interval, negative T wave in lateral leads, abnormal T wave axis, abnormal left or right intrinsicoid deflection duration, low voltage, left bundle branch block, right bundle branch block, left posterior hemiblock, left or right axis deviation, left ventricular hypertrophy, right or left atrial enlargement and pathological q(Q) wave in either group.
CONCLUSION: The study showed no direct effect of earthquake related PTSD on surface ECG in young patients. So, we propose that PTSD has no direct effect on surface ECG but may cause electrocardiographic changes indirectly by triggering atherosclerosis and/or contributing to the ongoing atherosclerotic process.
Collapse
|
11
|
Gao K, Li X, Zhang L, Bai L, Dong W, Gao K, Shi G, Xia X, Wu L, Zhang L. Transgenic expression of IL-33 activates CD8(+) T cells and NK cells and inhibits tumor growth and metastasis in mice. Cancer Lett 2013; 335:463-71. [PMID: 23499895 DOI: 10.1016/j.canlet.2013.03.002] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Revised: 02/27/2013] [Accepted: 03/04/2013] [Indexed: 12/29/2022]
Abstract
IL-33 is a multifunctional cytokine in immune regulation that activates Th1 cells, Th2 cells, CD8(+) T cells and NK cells. Our study showed that transgenic expression of IL-33 attenuated tumor metastasis in the B16 melanoma and Lewis lung carcinoma (LLC) metastatic models. The percentages and cytotoxicity of CD8(+) T cells and NK cells and their infiltration into the tumor tissues were significantly increased by the transgenic expression of IL-33 in tumor-bearing mice. Treatment with recombinant IL-33 could also increase the cytotoxicity of CD8(+) T cells and NK cells in vitro. In addition, depletion of CD8(+) T cells and NK cells using anti-CD8 or anti-asialo GM1 antibody abolished the pulmonary metastasis inhibition mediated by IL-33. Furthermore, IL-33 stimulated the activation of NF-κB and increased CD69 expression, which is a marker of the activated form of the two cell subsets, in CD8(+) T cells and NK cells. Our results suggest that IL-33 stimulated NF-κB signaling and promoted the proliferation, activation and infiltration of CD8(+) T cells and NK cells, which resulted in the inhibition of pulmonary metastasis in B16 melanoma and LLC mice models.
Collapse
Affiliation(s)
- Kun Gao
- Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, and Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Peking Union Medical College, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Philipson CW, Bassaganya-Riera J, Viladomiu M, Pedragosa M, Guerrant RL, Roche JK, Hontecillas R. The role of peroxisome proliferator-activated receptor γ in immune responses to enteroaggregative Escherichia coli infection. PLoS One 2013; 8:e57812. [PMID: 23469071 PMCID: PMC3585146 DOI: 10.1371/journal.pone.0057812] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Accepted: 01/25/2013] [Indexed: 12/24/2022] Open
Abstract
Background Enteroaggregative Escherichia coli (EAEC) is recognized as an emerging cause of persistent diarrhea and enteric disease worldwide. Mucosal immunity towards EAEC infections is incompletely understood due in part to the lack of appropriate animal models. This study presents a new mouse model and investigates the role of peroxisome proliferator-activated receptor gamma (PPARγ) in the modulation of host responses to EAEC in nourished and malnourished mice. Methods/Principal Findings Wild-type and T cell-specific PPARγ null C57BL/6 mice were fed protein-deficient diets at weaning and challenged with 5×109cfu EAEC strain JM221 to measure colonic gene expression and immune responses to EAEC. Antigen-specific responses to E. coli antigens were measured in nourished and malnourished mice following infection and demonstrated the immunosuppressive effects of malnutrition at the cellular level. At the molecular level, both pharmacological blockade and deletion of PPARγ in T cells resulted in upregulation of TGF-β, IL-6, IL-17 and anti-microbial peptides, enhanced Th17 responses, fewer colonic lesions, faster clearance of EAEC, and improved recovery. The beneficial effects of PPARγ blockade on weight loss and EAEC clearance were abrogated by neutralizing IL-17 in vivo. Conclusions Our studies provide in vivo evidence supporting the beneficial role of mucosal innate and effector T cell responses on EAEC burden and suggest pharmacological blockade of PPARγ as a novel therapeutic intervention for EAEC infection.
Collapse
Affiliation(s)
- Casandra W. Philipson
- Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, United States of America
- Center for Modeling Immunity to Enteric Pathogens, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Josep Bassaganya-Riera
- Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, United States of America
- Center for Modeling Immunity to Enteric Pathogens, Virginia Tech, Blacksburg, Virginia, United States of America
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Monica Viladomiu
- Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, United States of America
- Center for Modeling Immunity to Enteric Pathogens, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Mireia Pedragosa
- Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, United States of America
- Center for Modeling Immunity to Enteric Pathogens, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Richard L. Guerrant
- Division of Infectious Diseases and International Health, Center for Global Health, University of Virginia, Charlottesville, Virginia, United States of America
| | - James K. Roche
- Division of Infectious Diseases and International Health, Center for Global Health, University of Virginia, Charlottesville, Virginia, United States of America
| | - Raquel Hontecillas
- Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, United States of America
- Center for Modeling Immunity to Enteric Pathogens, Virginia Tech, Blacksburg, Virginia, United States of America
- * E-mail:
| |
Collapse
|
13
|
Pre-clinical studies of Notch signaling inhibitor RO4929097 in inflammatory breast cancer cells. Breast Cancer Res Treat 2012; 134:495-510. [PMID: 22547109 DOI: 10.1007/s10549-012-2075-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Accepted: 04/17/2012] [Indexed: 02/06/2023]
Abstract
Basal breast cancer, common among patients presenting with inflammatory breast cancer (IBC), has been shown to be resistant to radiation and enriched in cancer stem cells. The Notch pathway plays an important role in self-renewal of breast cancer stem cells and contributes to inflammatory signaling which promotes the breast cancer stem cell phenotype. Herein, we inhibited Notch signaling using a gamma secretase inhibitor, RO4929097, in an in vitro model that enriches for cancer initiating cells (3D clonogenic assay) and conventional 2D clonogenic assay to compare the effect on radiosensitization of the SUM149 and SUM190 IBC cell lines. RO4929097 downregulated the Notch target genes Hes1, Hey1, and HeyL, and showed a significant reduction in anchorage independent growth in SUM190 and SUM149. However, the putative self-renewal assay mammosphere formation efficiency was increased with the drug. To assess radiosensitization of putative cancer stem cells, cells were exposed to increasing doses of radiation with or without 1 μM RO4929097 in their standard (2D) and self-renewal enriching (3D) culture conditions. In the conventional 2D clonogenic assay, RO4929097 significantly sensitized SUM190 cells to ionizing radiation and has a modest radiosensitization effect in SUM149 cells. In the 3D clonogenic assays, however, a radioprotective effect was seen in both SUM149 and SUM190 cells at higher doses. Both cell lines express IL-6 and IL-8 cytokines known to mediate the efficacy of Notch inhibition and to promote self-renewal of stem cells. We further showed that RO429097 inhibits normal T-cell synthesis of some inflammatory cytokines, including TNF-α, a potential mediator of IL-6 and IL-8 production in the microenvironment. These data suggest that additional targeting agents may be required to selectively target IBC stem cells through Notch inhibition, and that evaluation of microenvironmental influences may shed further light on the potential effects of this inhibitor.
Collapse
|