1
|
Yang P, Liu Q, Zhang H, Wu M, Zhao J, Shen G, Zhao Y. Risk relationship between six autoimmune diseases and malignancies: An umbrella review. Autoimmun Rev 2025; 24:103779. [PMID: 39983807 DOI: 10.1016/j.autrev.2025.103779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 02/23/2025]
Abstract
BACKGROUND The bidirectional relationship between autoimmune diseases and malignancy has been widely discussed. And the relationship between autoimmune diseases and the risk of malignancy varies. Here, we categorized and re-analyzed the evidence of the association between six autoimmune diseases and malignancy risk, in order to provide ideas for the prevention of malignancy in the long-term individualized management of patients with autoimmune diseases. METHODS We systematically searched the relevant literatures in PubMed, Web of Science and Cochrane Library to identify and re-analyze studies methodically on the association between six autoimmune diseases and their malignancy risk. Our results showed that. RESULTS We included 34 meta-analyses including systematic lupus erythematosus, rheumatoid arthritis, psoriasis, ankylosing spondylitis, primary Sjogren's syndrome, multiple sclerosis, totalling 742 studies. Our results showed that the remaining five AIDs, with the exception of MS, were positively associated with the risk of overall malignancy. Among them, patients with SLE had the highest risk of developing lymphomas, oropharyngeal cancer and non-Hodgkin's lymphoma, and the lowest risk of developing uterine cancer, melanoma and endometrial cancer. The RA patients had the highest risk of developing lymphomas, Hodgkin's lymphoma and non-Hodgkin's and the lowest risk of colon cancer. pSS patients had the highest risk of lymphoma. MS patients had the highest risk of lung cancer and the lowest risk of testicular cancer. AS patients had the highest risk of lymphoblastic leukemia. PsO patients had the highest risk of keratinocyte cancer. CONCLUSION Patients with systematic lupus erythematosus, rheumatoid arthritis, psoriasis, ankylosing spondylitis and primary Sjogren's syndrome lead to an increased risk of overall malignancy, whereas patients with MS lead to a decreased risk of overall malignancy. However, the risk relationship between the same AIDs and different malignancies varied.
Collapse
Affiliation(s)
- Ping Yang
- Qinghai University, China; The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, China
| | | | - Hengheng Zhang
- Qinghai University, China; The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, China
| | - Meijie Wu
- Qinghai University, China; The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, China
| | - Jiuda Zhao
- The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, China
| | - Guoshuang Shen
- The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, China
| | - Yi Zhao
- The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, China.
| |
Collapse
|
2
|
Toumi E, Bestion E, Militello M, Lepidi H, Plauzolles A, Bardin N, Bertin D, Chiche L, Mege JL, Halfon P, Mezouar S. GNS561 (ezurpimtrostat), a small basic lipophilic molecule, prevents lupus phenotype in a pristane-induced lupus mouse model. Br J Pharmacol 2025. [PMID: 40258389 DOI: 10.1111/bph.70058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/23/2025] Open
Abstract
BACKGROUND AND PURPOSE Systemic lupus erythematosus is an autoimmune, multisystemic disease affecting all organs in the body. Accrued evidence has elucidated a role for autophagy in the onset and severity of systemic lupus erythematosus. The antimalarial drug hydroxychloroquine constitutes the cornerstone of standard of care for systemic lupus erythematosus, together with glucocorticoids and immunosuppressants or biologics, and all accompanied by various side effects. EXPERIMENTAL APPROACH AND OBJECTIVE Our study aimed to investigate the potential of GNS561 (ezurpimtrostat) treatment, a small basic lipophilic molecule that induces lysosomal dysregulation, using the pristane-induced lupus mouse model. KEY RESULTS Compared with hydroxychloroquine, GNS561 treatment presents a more pronounced impact on the development of pathogenic anti-antibodies in pristane-induced lupus mice. Next, focussing on clinical impact, we showed that GNS561 significantly reduced clinical signs of lupus in pristane-induced lupus by preventing the incidence and severity of arthritis, occurrence of nephritis and lung damage. Finally, GNS561 modulated the inflammatory profile in pristane-induced lupus mice through a reduction of the lipogranuloma score. Interestingly, focussing on interferon-α, only pristane-induced lupus mice treated by GNS561 presented a significant decrease of the cytokine, suggesting a higher efficacy for GNS561 in the modulation of lupus-induced inflammation compared with hydroxychloroquine. CONCLUSION All results show that GNS561, but not hydroxychloroquine, represents as an effective treatment to prevent clinical and inflammatory signs of lupus in this mouse model. IMPLICATIONS Altogether, this study highlights GNS561 as a promising investigational drug for systemic lupus erythematosus.
Collapse
Affiliation(s)
- Eya Toumi
- Aix-Marseille Univ, Microbe, Evolution, Phylogenie et Infection, Assistance Publique- Hopitaux de Marseille, Marseille, France
- Laboratoire Alphabio, Marseille, France
| | | | - Muriel Militello
- Aix-Marseille Univ, Microbe, Evolution, Phylogenie et Infection, Assistance Publique- Hopitaux de Marseille, Marseille, France
| | - Hubert Lepidi
- Aix-Marseille Univ, Microbe, Evolution, Phylogenie et Infection, Assistance Publique- Hopitaux de Marseille, Marseille, France
| | | | - Nathalie Bardin
- Aix Marseille Univ, Institut National de la Santé et de la Recherche Médicale, Centre de recherche en CardioVasculaire et Nutrition, Marseille, France
- Service d'Immunologie, Biogénopôle, Hôpital de la Timone, Assistance Publique-Hôpitaux de Marseille, Marseille, France
| | - Daniel Bertin
- Service d'Immunologie, Biogénopôle, Hôpital de la Timone, Assistance Publique-Hôpitaux de Marseille, Marseille, France
| | - Laurent Chiche
- Unité de Médecine Interne et Recherche Clinique, Hôpital Européen Marseille, Marseille, France
| | - Jean-Louis Mege
- Service d'Immunologie, Biogénopôle, Hôpital de la Timone, Assistance Publique-Hôpitaux de Marseille, Marseille, France
- Aix-Marseille Univ, Centre National de la Recherche Scientifique, Établissement Français du Sang, Anthropologie bio-culturelle, Droit, Éthique et Santé, Marseille, France
| | - Philippe Halfon
- Aix-Marseille Univ, Microbe, Evolution, Phylogenie et Infection, Assistance Publique- Hopitaux de Marseille, Marseille, France
- Laboratoire Alphabio, Marseille, France
- Genoscience Pharma, Marseille, France
- Infectious and Internal Medicine Department, Hôpital Européen Marseille, Marseille, France
| | - Soraya Mezouar
- Aix-Marseille Univ, Centre National de la Recherche Scientifique, Établissement Français du Sang, Anthropologie bio-culturelle, Droit, Éthique et Santé, Marseille, France
- Faculty of Medical and Paramedical Sciences, Aix-Marseille University, HIPE Human Lab, Marseille, France
| |
Collapse
|
3
|
Michalak KP, Michalak AZ. Understanding chronic inflammation: couplings between cytokines, ROS, NO, Ca i 2+, HIF-1α, Nrf2 and autophagy. Front Immunol 2025; 16:1558263. [PMID: 40264757 PMCID: PMC12012389 DOI: 10.3389/fimmu.2025.1558263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/14/2025] [Indexed: 04/24/2025] Open
Abstract
Chronic inflammation is an important component of many diseases, including autoimmune diseases, intracellular infections, dysbiosis and degenerative diseases. An important element of this state is the mainly positive feedback between inflammatory cytokines, reactive oxygen species (ROS), nitric oxide (NO), increased intracellular calcium, hypoxia-inducible factor 1-alpha (HIF-1α) stabilisation and mitochondrial oxidative stress, which, under normal conditions, enhance the response against pathogens. Autophagy and the nuclear factor erythroid 2-related factor 2 (Nrf2)-mediated antioxidant response are mainly negatively coupled with the above-mentioned elements to maintain the defence response at a level appropriate to the severity of the infection. The current review is the first attempt to build a multidimensional model of cellular self-regulation of chronic inflammation. It describes the feedbacks involved in the inflammatory response and explains the possible pathways by which inflammation becomes chronic. The multiplicity of positive feedbacks suggests that symptomatic treatment of chronic inflammation should focus on inhibiting multiple positive feedbacks to effectively suppress all dysregulated elements including inflammation, oxidative stress, calcium stress, mito-stress and other metabolic disturbances.
Collapse
Affiliation(s)
- Krzysztof Piotr Michalak
- Laboratory of Vision Science and Optometry, Physics and Astronomy Faculty, Adam Mickiewicz University in Poznań, Poznań, Poland
| | | |
Collapse
|
4
|
Zhang S, Hu W, Tang Y, Chen X. Identification and validation of key autophagy-related genes in lupus nephritis by bioinformatics and machine learning. PLoS One 2025; 20:e0318280. [PMID: 39869603 PMCID: PMC11771862 DOI: 10.1371/journal.pone.0318280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 01/13/2025] [Indexed: 01/29/2025] Open
Abstract
INTRODUCTION Lupus nephritis (LN) is one of the most frequent and serious organic manifestations of systemic lupus erythematosus (SLE). Autophagy, a new form of programmed cell death, has been implicated in a variety of renal diseases, but the relationship between autophagy and LN remains unelucidated. METHODS We analyzed differentially expressed genes (DEGs) in kidney tissues from 14 LN patients and 7 normal controls using the GSE112943 dataset. Key modules and their contained genes were identified utilizing weighted gene co-expression network analysis (WGCNA). Differentially expressed autophagy-related genes (DE-ARGs) among DEGs, key module genes and autophagy-related genes (ARGs) were obtained by venn plot, and subjected to protein-protein interaction network construction. Two machine learning methods were applied to identify signature genes. The area under the receiver operating characteristic (ROC) curves was used to assess the accuracy of the signature genes. We also analyzed immune cell infiltration in LN. Additionally, the association between key genes and kidney diseases was predicted. Finally, key genes expression in kidney was verified by clinical samples and animal experiments. RESULTS A total of 10304 DEGs were identified in GSE1129943 and 29 modules were identified in WGCNA. Among them, the brown module and coral 2 module exhibited significant correlation with LN (cor = 0.86, -0.84, p<0.001). Machine learning techniques identified 5 signature genes, but only 2 were validated in the external dataset GSE32591, namely MAP1LC3B (AUC = 0.920) and TNFSF10 (AUC = 0.937), which are involved in autophagy and apoptosis. Immune infiltration analysis suggested that these key genes may be associated with immune cell infiltration in LN. In addition, these genes have been linked to a variety of renal diseases, and their expression was verified in kidney tissues in LN patients and lupus mice. CONCLUSION MAP1LC3B and TNFSF10 may be key autophagy-related genes in LN. These key genes have the potential to provide new insights into the molecular diagnosis and treatment of LN.
Collapse
Affiliation(s)
- Su Zhang
- Department of Rheumatology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, P.R. China
| | - Weitao Hu
- Department of Rheumatology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, P.R. China
| | - Yelin Tang
- General Hospital of Ningxia Medical University, Yinchuan, P.R. China
| | - Xiaoqing Chen
- Department of Rheumatology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, P.R. China
| |
Collapse
|
5
|
Lee HK, Kim HK, Kim JY, Kim JS, Park J, Kim MS, Lee TY, Lim KH, Park H, Son DJ, Hong JT, Han SB. Ingenol-3-Angelate Enhances the B Cell Inhibitory Potential of Mesenchymal Stem Cells, Leading to Marked Alleviation of Lupus Symptoms in MRL. faslpr Mice. Int J Mol Sci 2024; 25:12625. [PMID: 39684336 DOI: 10.3390/ijms252312625] [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: 10/08/2024] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by autoantibody production by hyper-activated B cells. Although mesenchymal stem cells (MSCs) relieve lupus symptoms by inhibiting mainly T cells, whether MSCs also inhibit B cells has been controversial. Here, we found that naïve MSCs inhibited IFN-γ production by T cells, but not IgM production by B cells. We used a chemical approach to prime MSCs to inhibit B cells. We found that ingenol-3-angelate (I3A), a non-tumor-promoting phorbol ester, activated MSCs to inhibit B cells in a TGF-β1-dependent manner. We also showed that IL-1β induced MSCs to continuously secrete TGF-β1, which directly inhibited IgM production by B cells, whereas IL-1β did not. I3A-treated MSCs were better than naïve MSCs at ameliorating SLE symptoms in MRL.faslpr mice. In summary, our data provide information on how to generate MSCs that are effective for the treatment of SLE characterized by excessive B cell activation.
Collapse
Affiliation(s)
- Hong Kyung Lee
- College of Pharmacy, Chungbuk National University, Chungbuk 28160, Republic of Korea
- Bioengineering Institute, CorestemChemon Inc., Gyeonggi 13486, Republic of Korea
| | - Hwa Kyung Kim
- College of Pharmacy, Chungbuk National University, Chungbuk 28160, Republic of Korea
| | - Ji Yeon Kim
- College of Pharmacy, Chungbuk National University, Chungbuk 28160, Republic of Korea
| | - Ji Su Kim
- College of Pharmacy, Chungbuk National University, Chungbuk 28160, Republic of Korea
| | - JinKyung Park
- College of Pharmacy, Chungbuk National University, Chungbuk 28160, Republic of Korea
- Bioengineering Institute, CorestemChemon Inc., Gyeonggi 13486, Republic of Korea
| | - Min Sung Kim
- College of Pharmacy, Chungbuk National University, Chungbuk 28160, Republic of Korea
- Bioengineering Institute, CorestemChemon Inc., Gyeonggi 13486, Republic of Korea
| | - Tae Yong Lee
- College of Pharmacy, Chungbuk National University, Chungbuk 28160, Republic of Korea
- Bioengineering Institute, CorestemChemon Inc., Gyeonggi 13486, Republic of Korea
| | - Key-Hwan Lim
- College of Pharmacy, Chungbuk National University, Chungbuk 28160, Republic of Korea
| | - Hanseul Park
- College of Pharmacy, Chungbuk National University, Chungbuk 28160, Republic of Korea
| | - Dong Ju Son
- College of Pharmacy, Chungbuk National University, Chungbuk 28160, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy, Chungbuk National University, Chungbuk 28160, Republic of Korea
| | - Sang-Bae Han
- College of Pharmacy, Chungbuk National University, Chungbuk 28160, Republic of Korea
| |
Collapse
|
6
|
Zhang S, Xu R, Kang L. Biomarkers for systemic lupus erythematosus: A scoping review. Immun Inflamm Dis 2024; 12:e70022. [PMID: 39364719 PMCID: PMC11450456 DOI: 10.1002/iid3.70022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 08/31/2024] [Accepted: 09/06/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND In recent years, newly discovered potential biomarkers have great research potential in the diagnosis, disease activity prediction, and treatment of systemic lupus erythematosus (SLE). OBJECTIVE In this study, a scoping review of potential biomarkers for SLE over several years has identified the extent to which studies on biomarkers for SLE have been conducted, the specificity, sensitivity, and diagnostic value of potential biomarkers of SLE, the research potential of these biomarkers in disease diagnosis, and activity detection is discussed. METHODS In PubMed and Google Scholar databases, "SLE," "biomarkers," "predictor," "autoimmune diseases," "lupus nephritis," "neuropsychiatric SLE," "diagnosis," "monitoring," and "disease activity" were used as keywords to systematically search for SLE molecular biomarkers published from 2020 to 2024. Analyze and summarize the literature that can guide the article. CONCLUSIONS Recent findings suggest that some potential biomarkers may have clinical application prospects. However, to date, many of these biomarkers have not been subjected to repeated clinical validation. And no single biomarker has sufficient sensitivity and specificity for SLE. It is not scientific to choose only one or several biomarkers to judge the complex disease of SLE. It may be a good direction to carry out a meta-analysis of various biomarkers to find SLE biomarkers suitable for clinical use, or to evaluate SLE by combining multiple biomarkers through mathematical models. At the same time, advanced computational methods are needed to analyze large data sets and discover new biomarkers, and strive to find biomarkers that are sensitive and specific enough to SLE and can be used in clinical practice, rather than only staying in experimental research and data analysis.
Collapse
Affiliation(s)
- Su‐jie Zhang
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, Key Laboratory of High Altitude Environment and Genes Related to Diseases of Tibet Autonomous RegionSchool of Medicine, Xizang Minzu UniversityXianyangShaanxiChina
| | - Rui‐yang Xu
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, Key Laboratory of High Altitude Environment and Genes Related to Diseases of Tibet Autonomous RegionSchool of Medicine, Xizang Minzu UniversityXianyangShaanxiChina
| | - Long‐li Kang
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, Key Laboratory of High Altitude Environment and Genes Related to Diseases of Tibet Autonomous RegionSchool of Medicine, Xizang Minzu UniversityXianyangShaanxiChina
| |
Collapse
|
7
|
Yin HQ, Li XF, Fu Y, Zhu HL, Luo YS. A case report and review of rheumatoid arthritis co-occurring with tuberous sclerosis complex, a rare occurrence. Front Immunol 2024; 15:1425988. [PMID: 39391300 PMCID: PMC11464351 DOI: 10.3389/fimmu.2024.1425988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 09/03/2024] [Indexed: 10/12/2024] Open
Abstract
Rheumatoid arthritis (RA) is a common autoimmune disease. Tuberous sclerosis complex(TSC) is a rare autosomal dominant disorder. We report a case of RA with TSC. The patient was a 46-year-old woman with polyarthritis and cough symptoms, rheumatoid arthritis associated interstitial lung disease (RA-ILD) was initially considered, and after more than 3 months of anti-rheumatic treatment, the patient still had cough, and further examination revealed that the patient had lymphangioleiomyomatosis in the lungs, hepatic and renal angiomyolipomas, multiple subependymal nodules, Vertebral osteosclerotic nodules, as well as facial angiofibromas and periungual fibroma, RA was finally diagnosed with TSC, and everolimus 10mg qd was added to anti-rheumatic therapy for 1 month, and the patient's cough symptoms were relieved.
Collapse
Affiliation(s)
- Hai-Qin Yin
- Department of Rheumatology and Immunology, Jiujiang University Affiliated Hospital, Jiujiang, Jiangxi, China
| | | | | | | | | |
Collapse
|
8
|
Cao S, Jiang J, Yin H, Wang L, Lu Q. Abnormal energy metabolism in the pathogenesis of systemic lupus erythematosus. Int Immunopharmacol 2024; 134:112149. [PMID: 38692019 DOI: 10.1016/j.intimp.2024.112149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/20/2024] [Accepted: 04/21/2024] [Indexed: 05/03/2024]
Abstract
Systemic lupus erythematosus (SLE) is a severe autoimmune disease with significant socioeconomic impact worldwide. Orderly energy metabolism is essential for normal immune function, and disordered energy metabolism is increasingly recognized as an important contributor to the pathogenesis of SLE. Disorders of energy metabolism are characterized by increased reactive oxygen species, ATP deficiency, and abnormal metabolic pathways. Oxygen and mitochondria are critical for the production of ATP, and both mitochondrial dysfunction and hypoxia affect the energy production processes. In addition, several signaling pathways, including mammalian target of rapamycin (mTOR)/adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) signaling and the hypoxia-inducible factor (HIF) pathway also play important regulatory roles in energy metabolism. Furthermore, drugs with clear clinical effects on SLE, such as sirolimus, metformin, and tacrolimus, have been proven to improve the disordered energy metabolism of immune cells, suggesting the potential of targeting energy metabolism for the treatment of SLE. Moreover, several metabolic modulators under investigation are expected to have potential therapeutic effects in SLE. This review aimed to gain insights into the role and mechanism of abnormal energy metabolism in the pathogenesis of SLE, and summarizes the progression of metabolic modulator in the treatment of SLE.
Collapse
Affiliation(s)
- Shumei Cao
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, 210042, China; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China
| | - Jiao Jiang
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, 210042, China
| | - Haoyuan Yin
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, 210042, China; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China
| | - Lai Wang
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, 210042, China; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China.
| | - Qianjin Lu
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, 210042, China; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China; Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| |
Collapse
|
9
|
Samavati SF, Yarani R, Kiani S, HoseinKhani Z, Mehrabi M, Levitte S, Primavera R, Chetty S, Thakor AS, Mansouri K. Therapeutic potential of exosomes derived from mesenchymal stem cells for treatment of systemic lupus erythematosus. J Inflamm (Lond) 2024; 21:20. [PMID: 38867277 PMCID: PMC11170788 DOI: 10.1186/s12950-024-00381-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 03/14/2024] [Indexed: 06/14/2024] Open
Abstract
Autoimmune diseases are caused by an imbalance in the immune system, producing autoantibodies that cause inflammation leading to tissue damage and organ dysfunction. Systemic Lupus Erythematosus (SLE) is one of the most common autoimmune diseases and a major contributor to patient morbidity and mortality. Although many drugs manage the disease, curative therapy remains elusive, and current treatment regimens have substantial side effects. Recently, the therapeutic potential of exosomes has been extensively studied, and novel evidence has been demonstrated. A direct relationship between exosome contents and their ability to regulate the immune system, inflammation, and angiogenesis. The unique properties of extracellular vesicles, such as biomolecule transportation, biodegradability, and stability, make exosomes a promising treatment candidate for autoimmune diseases, particularly SLE. This review summarizes the structural features of exosomes, the isolation/purification/quantification method, their origin, effect, immune regulation, a critical consideration for selecting an appropriate source, and their therapeutic mechanisms in SLE.
Collapse
Affiliation(s)
- Shima Famil Samavati
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Reza Yarani
- Translational Type 1 Diabetes Research, Department of Clinical Research, Steno Diabetes Center Copenhagen, Gentofte, Denmark
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Sara Kiani
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zohreh HoseinKhani
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Masomeh Mehrabi
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Steven Levitte
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Rosita Primavera
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Shashank Chetty
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Avnesh S Thakor
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Kamran Mansouri
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
10
|
Xing J, Wang K, Xu YC, Pei ZJ, Yu QX, Liu XY, Dong YL, Li SF, Chen Y, Zhao YJ, Yao F, Ding J, Hu W, Zhou RP. Efferocytosis: Unveiling its potential in autoimmune disease and treatment strategies. Autoimmun Rev 2024; 23:103578. [PMID: 39004157 DOI: 10.1016/j.autrev.2024.103578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 07/06/2024] [Accepted: 07/07/2024] [Indexed: 07/16/2024]
Abstract
Efferocytosis is a crucial process whereby phagocytes engulf and eliminate apoptotic cells (ACs). This intricate process can be categorized into four steps: (1) ACs release "find me" signals to attract phagocytes, (2) phagocytosis is directed by "eat me" signals emitted by ACs, (3) phagocytes engulf and internalize ACs, and (4) degradation of ACs occurs. Maintaining immune homeostasis heavily relies on the efficient clearance of ACs, which eliminates self-antigens and facilitates the generation of anti-inflammatory and immunosuppressive signals that maintain immune tolerance. However, any disruptions occurring at any of the efferocytosis steps during apoptosis can lead to a diminished efficacy in removing apoptotic cells. Factors contributing to this inefficiency encompass dysregulation in the release and recognition of "find me" or "eat me" signals, defects in phagocyte surface receptors, bridging molecules, and other signaling pathways. The inadequate clearance of ACs can result in their rupture and subsequent release of self-antigens, thereby promoting immune responses and precipitating the onset of autoimmune diseases such as systemic lupus erythematosus, rheumatoid arthritis, type 1 diabetes, and multiple sclerosis. A comprehensive understanding of the efferocytosis process and its implications can provide valuable insights for developing novel therapeutic strategies that target this process to prevent or treat autoimmune diseases.
Collapse
Affiliation(s)
- Jing Xing
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; School of pharmacy, Anhui Medical University, Hefei 230032, China
| | - Ke Wang
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Yu-Cai Xu
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; School of pharmacy, Anhui Medical University, Hefei 230032, China
| | - Ze-Jun Pei
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; School of pharmacy, Anhui Medical University, Hefei 230032, China
| | - Qiu-Xia Yu
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; School of pharmacy, Anhui Medical University, Hefei 230032, China
| | - Xing-Yu Liu
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; School of pharmacy, Anhui Medical University, Hefei 230032, China
| | - Ya-Lu Dong
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; School of pharmacy, Anhui Medical University, Hefei 230032, China
| | - Shu-Fang Li
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Yong Chen
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Ying-Jie Zhao
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Feng Yao
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Jie Ding
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Wei Hu
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China.
| | - Ren-Peng Zhou
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China.
| |
Collapse
|
11
|
Ding M, Jin L, Zhao J, Yang L, Cui S, Wang X, He J, Chang F, Shi M, Ma J, Song S, Jin H, Liu A. Add-on sirolimus for the treatment of mild or moderate systemic lupus erythematosus via T lymphocyte subsets balance. Lupus Sci Med 2024; 11:e001072. [PMID: 38351097 PMCID: PMC10868177 DOI: 10.1136/lupus-2023-001072] [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: 10/06/2023] [Accepted: 01/31/2024] [Indexed: 02/16/2024]
Abstract
OBJECTIVE The efficacy of sirolimus in treating severe or refractory systemic lupus erythematosus (SLE) has been confirmed by small-scale clinical trials. However, few studies focused on mild or moderate SLE. Therefore, in this study we elucidated clinical efficacy of add-on sirolimus in patients with mild or moderate SLE. METHODS Data of 17 consecutive patients with SLE were retrospectively collected. SLE Disease Activity Index-2000 (SLEDAI-2K), clinical manifestation, laboratory data and peripheral T lymphocyte subsets with cytokines were collected before and 6 months after sirolimus add-on treatment. T cell subsets were detected by flow cytometry and cytokines were determined by multiplex bead-based flow fluorescent immunoassay simultaneously. Twenty healthy controls matched with age and sex were also included in our study. RESULTS (1) The numbers of peripheral blood lymphocytes, T cells, T helper (Th) cells, regulatory T (Treg) cells, Th1 cells, Th2 cells and Treg/Th17 ratios in patients with SLE were significantly lower, while the numbers of Th17 cells were evidently higher than those of healthy control (p<0.05). (2) After 6 months of sirolimus add-on treatment, urinary protein, pancytopenia, immunological indicators and SLEDAI-2K in patients with SLE were distinctively improved compared with those before sirolimus treatment (p<0.05). (3) The numbers of peripheral blood lymphocytes, T cells, Th cells, Treg cells, Th2 cells and the ratios of Treg/Th17 in patients with SLE after treatment were clearly higher than those before (p<0.05). (4) The levels of plasma interleukin (IL)-5, IL-6 and IL-10 in patients with SLE decreased notably, conversely the IL-4 levels increased remarkably compared with pretreatment (p<0.05). CONCLUSIONS (1) Patients with SLE presented imbalanced T cell subsets, especially the decreased ratio of Treg/Th17. (2) Sirolimus add-on treatment ameliorated clinical involvement, serological abnormalities and disease activity without adverse reactions in patients with SLE. (3) The multi-target therapy facilitates the enhanced numbers of Treg cells, Treg/Th17 imbalance and anti-inflammatory cytokines, simultaneously, reducing inflammatory cytokines.
Collapse
Affiliation(s)
- Meng Ding
- Department of Rheumatology and Immunology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Lu Jin
- Department of Rheumatology and Immunology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jinwen Zhao
- Department of Rheumatology and Immunology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Lin Yang
- Department of Rheumatology and Immunology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shaoxin Cui
- Department of Rheumatology and Immunology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaoping Wang
- Department of Rheumatology and Immunology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jingjing He
- Department of Rheumatology and Immunology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Fei Chang
- Department of Rheumatology and Immunology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Min Shi
- Department of Clinical Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Laboratory Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jun Ma
- Department of Anatomy, Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, Hebei, China
| | - Shuran Song
- Department of Clinical Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Laboratory Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Hongtao Jin
- Department of Rheumatology and Immunology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Aijing Liu
- Department of Rheumatology and Immunology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Laboratory Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, Hebei, China
| |
Collapse
|
12
|
Stergioti EM, Manolakou T, Sentis G, Samiotaki M, Kapsala N, Fanouriakis A, Boumpas DT, Banos A. Transcriptomic and proteomic profiling reveals distinct pathogenic features of peripheral non-classical monocytes in systemic lupus erythematosus. Clin Immunol 2023; 255:109765. [PMID: 37678715 DOI: 10.1016/j.clim.2023.109765] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 09/04/2023] [Indexed: 09/09/2023]
Abstract
Peripheral blood monocytes propagate inflammation in systemic lupus erythematosus (SLE). Three major populations of monocytes have been recognized namely classical (CM), intermediate (IM) and non-classical monocytes (NCM). Herein, we performed a comprehensive transcriptomic, proteomic and functional characterization of the three peripheral monocytic subsets from active SLE patients and healthy individuals. Our data demonstrate extensive molecular disruptions in circulating SLE NCM, characterized by enhanced inflammatory features such as deregulated DNA repair, cell cycle and heightened IFN signaling combined with differentiation and developmental cues. Enhanced DNA damage, elevated expression of p53, G0 arrest of cell cycle and increased autophagy stress the differentiation potential of NCM in SLE. This immunogenic profile is associated with an activated macrophage phenotype of NCM exhibiting M1 characteristics in the circulation, fueling the inflammatory response. Together, these findings identify circulating SLE NCM as a pathogenic cell type in the disease that could represent an additional therapeutic target.
Collapse
Affiliation(s)
- Eirini Maria Stergioti
- Laboratory of Autoimmunity and Inflammation, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, Athens 115 27, Greece; 4th Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens Medical School, Athens 124 62, Greece.
| | - Theodora Manolakou
- Laboratory of Autoimmunity and Inflammation, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, Athens 115 27, Greece
| | - George Sentis
- Laboratory of Autoimmunity and Inflammation, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, Athens 115 27, Greece
| | - Martina Samiotaki
- Institute for Bioinnovation, Biomedical Sciences Research Center Alexander Fleming, Vari, Athens 166 72, Greece
| | - Noemin Kapsala
- 4th Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens Medical School, Athens 124 62, Greece
| | - Antonis Fanouriakis
- 4th Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens Medical School, Athens 124 62, Greece
| | - Dimitrios T Boumpas
- Laboratory of Autoimmunity and Inflammation, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, Athens 115 27, Greece.
| | - Aggelos Banos
- Laboratory of Autoimmunity and Inflammation, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, Athens 115 27, Greece.
| |
Collapse
|
13
|
Gao Y, Zhou J, Huang Y, Wang M, Zhang Y, Zhang F, Gao Y, Zhang Y, Li H, Sun J, Xie Z. Jiedu-Quyu-Ziyin Fang (JQZF) inhibits the proliferation and activation of B cells in MRL/lpr mice via modulating the AKT/mTOR/c-Myc signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2023:116625. [PMID: 37236380 DOI: 10.1016/j.jep.2023.116625] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/02/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Jiedu-Quyu-Ziyin Fang (JQZF) is a new herbal formula improved based on "Sheng Ma Bie Jia Tang" in the Golden Chamber, has been proved to be effective in the treatment of SLE. The ability of JQZF to prevent lymphocyte growth and survival has been demonstrated in earlier investigations. However, the specific mechanism of JQZF on SLE has not been fully investigated. AIM OF THE STUDY To reveal the potential mechanisms of JQZF inhibiting B cell proliferation and activation in MRL/lpr mice. MATERIALS AND METHODS MRL/lpr mice were treated with low-dose, high-dose JQZF and normal saline for 6 weeks. The effect of JQZF on disease improvement in MRL/lpr mice was studied using enzyme-linked immunosorbent assay (ELISA), histopathological staining, serum biochemical parameters and urinary protein levels. The changes of B lymphocyte subsets in the spleen were analyzed by flow cytometry. The contents of ATP and PA in B lymphocytes from the spleens of mice were determined by ATP content assay kit and PA assay kit. Raji cells (a B lymphocyte line) were selected as the cell model in vitro. The effects of JQZF on the proliferation and apoptosis of B cells were detected by flow cytometry and CCK8. The effect of JQZF on the AKT/mTOR/c-Myc signaling pathway in B cells were detected via western blot. RESULTS JQZF, especially at high dose, significantly improved the disease development of MRL/lpr mice. Flow cytometry results showed that JQZF affected the proliferation and activation of B cells. In addition, JQZF inhibited the production of ATP and PA in B lymphocytes. In vitro cell experiments further confirmed that JQZF can inhibit Raji proliferation and promote cell apoptosis through AKT/mTOR/c-Myc signaling pathway. CONCLUSION JQZF may affect the proliferation and activation of B cells by inhibiting the AKT/mTOR/c-Myc signaling pathway.
Collapse
Affiliation(s)
- YiNi Gao
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - JiaWang Zhou
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Yao Huang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - MeiJiao Wang
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Yi Zhang
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - FengQi Zhang
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Yan Gao
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - YiYang Zhang
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - HaiChang Li
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Jing Sun
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China.
| | - ZhiJun Xie
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
14
|
Lin W, Lin Z, Lin X, Peng Z, Liang X, Wei S. Integrated analysis and clinical correlation analysis of hub genes, immune infiltration, and potential therapeutic agents related to lupus nephritis. Lupus 2023; 32:633-643. [PMID: 36912500 DOI: 10.1177/09612033231161587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2023]
Abstract
BACKGROUND Lupus nephritis (LN) is the most common complication of systemic lupus erythematosus (SLE). This study aimed to explore biomarkers, mechanisms, and potential novel agents regarding LN through bioinformatic analysis. METHOD Four expression profiles were downloaded from the Gene Expression Omnibus (GEO) database and differentially expressed genes (DEGs) were acquired. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGGs) pathway enrichment analyses of DEGs were performed using the R software. The protein-protein interaction (PPI) network was developed using the STRING database. Additionally, five algorithms were used to screen out the hub genes. Expression of the hub genes were validated using Nephroseq v5. CIBERSORT was used to evaluate the infiltration of immune cells. Finally, The Drug-Gene Interaction Database was used to predict potential targeted drugs. RESULT FOS and IGF1 were identified as hub genes, with excellent specificity and sensitivity diagnosis of LN. FOS was also related to renal injury. LN patients had lower activated and resting dendritic cells (DCs) and higher M1 macrophages and activated NK cells than healthy control (HC). FOS had a positive correlation with activated mast cells and a negative correlation with resting mast cells. IGF1 had a positive correlation with activated DCs and a negative correlation with monocytes. The targeted drugs were dusigitumab and xentuzumab target for IGF1. CONCLUSION We analyzed the transcriptomic signature of LN along with the landscape of the immune cell. FOS and IGF1 are promising biomarkers for diagnosing and evaluating the progression of LN. The drug-gene interaction analyses provide a list of candidate drugs for the precise treatment of LN.
Collapse
Affiliation(s)
- Weiyi Lin
- Zhujiang Hospital, The Second School of Clinical Medicine, 70570Southern Medical University, Guangzhou, The People's Republic of China
| | - Zien Lin
- Zhujiang Hospital, The Second School of Clinical Medicine, 70570Southern Medical University, Guangzhou, The People's Republic of China
| | - Xiaobing Lin
- Zhujiang Hospital, The Second School of Clinical Medicine, 70570Southern Medical University, Guangzhou, The People's Republic of China
| | - Zhishen Peng
- Zhujiang Hospital, The Second School of Clinical Medicine, 70570Southern Medical University, Guangzhou, The People's Republic of China
| | - Xiaofeng Liang
- Zhujiang Hospital, The Second School of Clinical Medicine, 70570Southern Medical University, Guangzhou, The People's Republic of China
| | - Shanshan Wei
- Department of Dermatology, Zhujiang Hospital, 70570Southern Medical University, Guangzhou, The People's Republic of China
| |
Collapse
|
15
|
Ni FF, Liu GL, Jia SL, Li CR, Gao XJ. Effects of the mTOR Pathway on the Balance of Th2/Treg Cells in Children with Idiopathic Nephrotic Syndrome. Indian J Nephrol 2023; 33:93-100. [PMID: 37234433 PMCID: PMC10208544 DOI: 10.4103/ijn.ijn_521_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 05/19/2022] [Accepted: 05/29/2022] [Indexed: 05/28/2023] Open
Abstract
Introduction Immune dysfunction contributes to the progression of idiopathic nephrotic syndrome (INS), but the details of the pathogenesis of progression remain unknown. This study of children with INS investigated the relationship of activation of the mechanistic target of rapamycin (mTOR) pathway (PI3K/AKT/mTOR/p70S6K) with the levels of T helper 2/regulatory T (Th2/Treg) cells. Materials and Methods Twenty children with active INS (before steroid treatment), 20 children with remitting INS (INS-R, after steroid treatment), and 20 healthy control children (Ctrl) were enrolled. The levels of Th2/Treg cells in their peripheral circulatory systems were measured using flow cytometry, and the concentration of interleukin (IL)-4 was determined using a cytometric bead array (CBA). The levels of PI3K, AKT, mTOR, p70S6K, and transcription factors associated with Th2/Treg cells were measured using real-time polymerase chain reaction. Results The INS group had a greater proportion of circulating Th2 cells; level of IL-4 protein; and levels of GATA, PI3K, AKT, mTOR, and p70S6K mRNAs than the Ctrl group (all P < 0.05), but a lower proportion of circulating Tregs and expression of Foxp3 (both P < 0.05). Patients in the INS-R group had normalization of these markers (all P < 0.05). Patients in the INS group had negative correlation in the percentage of Treg cells with Th2 cells and with IL-4 level and a negative correlation in the levels of GATA3 and Foxp3 mRNAs. Conclusions Patients with active INS had an imbalance of Th2/Treg cells, which might result from the aberrant signaling of the mTOR pathway (PI3K/AKT/mTOR/p70S6K).
Collapse
Affiliation(s)
- Fen Fen Ni
- Department of Nephrology, Shenzhen Children’s Hospital, Shenzhen, China
| | - Guang Lei Liu
- Department of Pediatrics, The Fifth Affiliated (Zhuhai) Hospital of Zunyi Medical University, Zhuhai, China
| | - Shi Lei Jia
- Department of Nephrology, Shenzhen Children’s Hospital, Shenzhen, China
| | - Cheng Rong Li
- Department of Nephrology, Shenzhen Children’s Hospital, Shenzhen, China
| | - Xiao Jie Gao
- Department of Nephrology, Shenzhen Children’s Hospital, Shenzhen, China
| |
Collapse
|
16
|
The role of non-coding RNA in lupus nephritis. Hum Cell 2023; 36:923-936. [PMID: 36840837 DOI: 10.1007/s13577-023-00883-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 02/16/2023] [Indexed: 02/26/2023]
Abstract
Systemic lupus erythematosus (SLE) is a common autoimmune disease with multiple manifestations. The renal implication, also called lupus nephritis (LN) is the most regular type of complication and results in adverse outcomes. Multiple studies revealed the importance of non-coding RNA in diseases, likewise observed in nephropathies, particularly LN. Long-non-coding RNA (lncRNA) is a group of RNA that are more than 200 nucleotides in length. And in circular RNA (circRNA), the head and tail of RNA are connected by a 3' → 5' phosphodiester bond. Both two types of non-coding RNA play important roles in LN pathogenesis through the competitive endogenous RNA (ceRNA) effect. LncRNAs and circRNAs can sponge miRNAs and consequently act on downstream signaling pathways, which are capable to influence various aspects of LN, including cell proliferation, inflammation, and oxidative stress. And lncRNAs and circRNAs have the potential to act as biomarkers to diagnose LN and distinguish whether SLE patients with LN or not. In the future, lncRNAs and circRNAs may be accessible therapeutic targets.
Collapse
|
17
|
Lan J, Zhu Y, Rao J, Liu L, Gong A, Feng F, Chen B, Huang J, Zhang Y, Chu L, Zhong H, Li L, Yan J, Li W, Xue C. MTOR gene polymorphism may be associated with microscopic polyangiitis susceptibility in a Guangxi population of China. Gene X 2023; 854:147101. [PMID: 36496178 DOI: 10.1016/j.gene.2022.147101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 11/18/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE Microscopic polyangiitis (MPA) onset is affected by genetic predisposition. Autophagy plays a certain role in antineutrophil cytoplasmic antibody-associated vasculitis developing. A key factor in autophagy regulating, the genetic polymorphism of MTOR gene is essential. The objective was to explore the associations between MTOR gene polymorphism and MPA susceptibility in a Guangxi population of China. METHODS A sum of 208 MPA cases and 209 healthy volunteers from Guangxi in this case-control study, four important single nucleotide polymorphism (SNP) loci of MTOR gene including rs3806317, rs1064261, rs1883965 and rs2295080 were examined. Multiplex polymerase chain reaction combined with high-throughput sequencing was performed. Subgroup analysis was evaluated by gender and ethnicity. Linkage disequilibrium and haplotype analysis were tested. Multi-SNPs interaction among mTOR signaling pathway was assessed. RESULTS For rs2295080, homozygous mutant GG genotype was associated with a decreased susceptibility of MPA in recessive model (OR = 0.38, 95%CI: 0.14-1.00, p = 0.040), particularly in the subgroup of female (OR = 0.16, 95%CI: 0.03-0.74, p = 0.006) and Han population (OR = 0.32, 95%CI: 0.10-1.00, p = 0.034). Individual carrying G allele was linked with decreasing MPA susceptibility in Han population of Guangxi (OR = 0.65, 95%CI: 0.44-0.97, p = 0.036). In haplotype analysis, the haplotype AAT was correlated with increasing susceptibility of MPA (OR = 1.347, 95%CI: 1.004-1.807, p = 0.046). Moreover, in the multi-SNPs interaction analysis, the six-locus model was identified as the best interaction model (p < 0.05). CONCLUSION These findings suggest that rs2295080 polymorphism of MTOR gene may be associated with MPA susceptibility in a Guangxi population of China and G allele might be an important protective factor.
Collapse
Affiliation(s)
- Jingjing Lan
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China
| | - Yan Zhu
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China; The First Affiliated Hospital, Department of Nephrology, Hengyang Medical School, University of South China, Hengyang, Hunan, P.R. China
| | - Jinlan Rao
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China
| | - Liu Liu
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China
| | - Aimei Gong
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China; Department of Nephrology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, Guangxi, P.R. China
| | - Fei Feng
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China
| | - Bingfang Chen
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China
| | - Junxia Huang
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China
| | - Yurong Zhang
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China
| | - Liepeng Chu
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China
| | - Huan Zhong
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China
| | - Lizhen Li
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China; Department of Nephrology, Hunan Research Institute of Geriatrics, The Peoples Hospital of Hunan Province, Changsha, Hunan, P.R. China
| | - Jinlian Yan
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China
| | - Wei Li
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China.
| | - Chao Xue
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China.
| |
Collapse
|
18
|
Yan Z, Chen Q, Xia Y. Oxidative Stress Contributes to Inflammatory and Cellular Damage in Systemic Lupus Erythematosus: Cellular Markers and Molecular Mechanism. J Inflamm Res 2023; 16:453-465. [PMID: 36761905 PMCID: PMC9907008 DOI: 10.2147/jir.s399284] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 01/18/2023] [Indexed: 02/05/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a multifactorial autoimmune disease with complex pathogenesis, the treatment of which relies exclusively on the use of immunosuppressants. Increased oxidative stress is involved in causing inflammatory and cellular defects in the pathogenesis of SLE. Various inflammatory and cellular markers including oxidative modifications of proteins, lipids, and DNA contribute to immune system dysregulation and trigger an aggressive autoimmune attack through molecular mechanisms like enhanced NETosis, mTOR pathway activation, and imbalanced T-cell differentiation. Accordingly, the detection of inflammatory and cellular markers is important for providing an accurate assessment of the extent of oxidative stress. Oxidative stress also reduces DNA methylation, thus allowing the increased expression of affected genes. As a result, pharmacological approaches targeting oxidative stress yield promising results in treating patients with SLE. The purpose of this review is to examine the involvement of oxidative stress in the pathogenesis and management of SLE.
Collapse
Affiliation(s)
- Zhu Yan
- Department of Dermatology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004, People’s Republic of China
| | - Qin Chen
- Department of Dermatology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004, People’s Republic of China
| | - Yumin Xia
- Department of Dermatology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004, People’s Republic of China,Correspondence: Yumin Xia, Department of Dermatology, The Second Affiliated Hospital of Xi’an Jiaotong University, 157 Xiwu Road, Xi’an, 710004, People’s Republic of China, Tel/Fax +86-29-87679969, Email
| |
Collapse
|
19
|
Zhao X, Wang S, Wang S, Xie J, Cui D. mTOR signaling: A pivotal player in Treg cell dysfunction in systemic lupus erythematosus. Clin Immunol 2022; 245:109153. [DOI: 10.1016/j.clim.2022.109153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/30/2022] [Accepted: 10/04/2022] [Indexed: 11/03/2022]
|
20
|
Legorreta-Haquet MV, Santana-Sánchez P, Chávez-Sánchez L, Chávez-Rueda AK. The effect of prolactin on immune cell subsets involved in SLE pathogenesis. Front Immunol 2022; 13:1016427. [PMID: 36389803 PMCID: PMC9650038 DOI: 10.3389/fimmu.2022.1016427] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/13/2022] [Indexed: 08/27/2023] Open
Abstract
The higher frequency of autoimmune diseases in the female population compared to males suggests that certain hormones, such as prolactin (PRL), play a role in determining the prevalence of autoimmunity in women, particularly during childbearing age. PRL can act not only as a hormone but also as a cytokine, being able to modulate immune responses. Hyperprolactinemia has been implicated in the pathogenesis of various autoimmune diseases where it may affect disease activity. One of the conditions where PRL has such a role is systemic lupus erythematosus (SLE). PRL regulates the proliferation and survival of both lymphoid and myeloid cells. It also affects the selection of T-cell repertoires by influencing the thymic microenvironment. In autoimmune conditions, PRL interferes with the activity of regulatory T cells. It also influences B cell tolerance by lowering the activation threshold of anergic B cells. The production of CD40L and cytokines, such as interleukin IL-6, are also promoted by PRL. This, in turn, leads to the production of autoantibodies, one of the hallmarks of SLE. PRL increases the cytotoxic activity of T lymphocytes and the secretion of proinflammatory cytokines. The production of proinflammatory cytokines, particularly those belonging to the type 1 interferon (IFN) family, is part of the SLE characteristic genetic signature. PRL also participates in the maturation and differentiation of dendritic cells, promoting the presentation of autoantigens and high IFNα secretion. It also affects neutrophil function and the production of neutrophil traps. Macrophages and dendritic cells can also be affected by PRL, linking this molecule to the abnormal behavior of both innate and adaptive immune responses.This review aimed to highlight the importance of PRL and its actions on the cells of innate and adaptive immune responses. Additionally, by elucidating the role of PRL in SLE etiopathogenesis, this work will contribute to a better understanding of the factors involved in SLE development and regulation.
Collapse
Affiliation(s)
| | | | | | - Adriana Karina Chávez-Rueda
- Unidad de Investigación Médica en Inmunología (UIM) en Inmunología, Hospital de Pediatría, Centro Médico Nacional (CMN) Siglo XXI, Instituto Mexicano del Seguro Social, México City, Mexico
| |
Collapse
|
21
|
Koh JS, Oh S, Chung C. Pulmonary lymphangioleiomyomatosis and renal angiomyolipoma in a patient with systemic lupus erythematosus: A case report. Medicine (Baltimore) 2022; 101:e30554. [PMID: 36197220 PMCID: PMC9509188 DOI: 10.1097/md.0000000000030554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND The co-incidence of systemic lupus erythematosus (SLE) and tuberous sclerosis with pulmonary lymphangioleiomyomatosis (LAM) and renal angiomyolipoma (AML) is rare. In such patients, the rupture of renal AML may result in fatal circumstances, but this may be preventable. METHODS A 22-year-old Asian woman with SLE was admitted to our hospital with severe left-flank pain. Imaging studies showed the bilateral rupture of multiple renal AMLs. RESULTS The patient underwent emergency selective transcatheter embolization (TE) of the left renal artery. After TE and massive hydration, the patient complained of dyspnea and postembolization syndrome with fever. The chest computed tomography (CT) revealed pulmonary LAM, pulmonary edema with bilateral pleural effusions, and pneumonic consolidation. After the emergency procedure, the patient was treated with intravenous administration of antibiotics, diuretics, and nonsteroidal anti-inflammatory drugs for 10 days. The patient recovered favorably and was discharged 20 days after the treatment. She was diagnosed with renal AML and pulmonary LAM along with facial angiofibromas as well as tuberous sclerosis complex (TSC), although she had no TSC1 or TSC2 gene mutations. CONCLUSION Although rare, SLE may coexist with TSC, along with LAM and AML, with a risk of AML rupture. The activation of the mTOR signaling pathway is shared between SLE and TSC. Thus, in patients with SLE, clinicians should consider imaging studies, such as kidney sonography and chest CT, to screen for possible manifestation of AML and LAM.
Collapse
Affiliation(s)
- Jeong Suk Koh
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon, 301-721, Republic of Korea
| | - Sina Oh
- College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Chaeuk Chung
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon, 301-721, Republic of Korea
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea
- * Correspondence: Chaeuk Chung, Division of Pulmonology, Department of Internal Medicine, Chungnam National University, 282, Munhwaro, Jung-gu, Daejeon 35015, Republic of Korea (e-mail: )
| |
Collapse
|
22
|
Exploring the Molecular Mechanism of Zhi Bai Di Huang Wan in the Treatment of Systemic Lupus Erythematosus Based on Network Pharmacology and Molecular Docking Techniques. Processes (Basel) 2022. [DOI: 10.3390/pr10101914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Objective: To investigate the molecular mechanism and simulated validation of Zhi Bai Di Huang Pill (ZBDHP) for the treatment of systemic lupus erythematosus (SLE) using network pharmacology and molecular docking techniques. Methods: The active ingredients of ZBDHP were obtained through the TCMSP database and the Canonical SMILES of the active ingredients were queried through Pubchem. The targets of the active ingredients were predicted in the SwissTarget database based on the SMILES. The SLE-related disease targets were obtained through the GeneCards, OMIM and DisGenets databases, and the intersection targets of ZBDHP and SLE were obtained using the Venny 2.1.0 online platform. Intersection targets build a visual protein interaction network (PPI) through the STRING database, and the core targets were identified by network topology analysis. GO analysis and KEGG pathway enrichment analysis of the intersecting targets were performed using the DAVID database. Finally, the molecular docking of the first four active ingredients and the first four core target genes were verified by Pubchem, the PDB database and CB-Dock online molecular docking technology. Results: ZBDHP screened 91 potential active ingredients and 816 potential targets. Among them, 141 genes were intersected by ZBDHP and SLE. The network topology analysis showed that the main active ingredients were Hydroxygenkwanin, Alisol B, asperglaucide, Cerevisterol, etc., and the key target genes were TNF, AKT1, EGFR, STAT3, etc. GO and KEGG enrichment analysis showed that common targets interfere with biological processes or molecular functions such as signal transduction protein phosphorylation, inflammatory response, transmembrane receptor protein tyrosine kinase activity, etc., through multiple signaling pathways, such as pathways in cancer, Kaposi sarcoma-associated herpesvirus infection, the PI3K-Akt signaling pathway, lipid and atherosclerosis, hepatitis B, etc. Molecular docking results showed that the active components of ZBDHP have good binding activity to the core targets of SLE. Conclusions: This study reveals that the ZBDHP treatment of SLE is a complex mechanistic process with multi-components, multi-targets and multi-pathways, and it may play a therapeutic role in SLE by inhibiting the production, proliferation and apoptosis of inflammatory factors. In conclusion, the present study provides a theoretical basis for further research on ZBDHP.
Collapse
|
23
|
Zhang X, Wang G, Bi Y, Jiang Z, Wang X. Inhibition of glutaminolysis ameliorates lupus by regulating T and B cell subsets and downregulating the mTOR/P70S6K/4EBP1 and NLRP3/caspase-1/IL-1β pathways in MRL/lpr mice. Int Immunopharmacol 2022; 112:109133. [PMID: 36113317 DOI: 10.1016/j.intimp.2022.109133] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/28/2022] [Accepted: 08/03/2022] [Indexed: 01/02/2023]
Abstract
BACKGROUND AND AIM OF THE STUDY Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by lymphocyte imbalance. The differentiation and function of T and B cells receive regulation from intracellular energy metabolism. Herein, we aimed to investigate glutamine metabolism levels in SLE and explore the effects of modulating glutamine metabolism on T and B cell subsets and related signaling pathways in MRL/lpr lupus mice. METHODS We assessed intracellular glutamine metabolism in SLE patients and MRL/lpr mice by measuring intracellular glutamate and Glutaminase 1 (GLS1) protein levels. Intraperitoneal injection of the GLS1 inhibitor CB839 was performed to reduce glutamine metabolism and lupus-like manifestations in MRL/lpr mice were evaluated. The proportions and numbers of T and B cell subsets were determinedvia flow cytometry. Pathway-related proteins were detected using western blotting. RESULTS In this study, we reported that glutamine metabolism levels were aberrantly elevated in splenic mononuclear cells from MRL/lpr lupus mice, as well as in peripheral blood mononuclear cells (PBMCs) of SLE patients. Inhibition of glutamine metabolism by CB839 treatment for 8 weeks alleviated the lupus-like manifestations in MRL/lpr mice, including the kidney lesions, urinary protein/creatinine ratio, spleen index, and serum IgG1. Meanwhile, CB839 treatment ameliorated the depletion of IL-10 producing B cells (B10) and adjusted the Th1/TH2 and TH17/Treg imbalance. The inhibition of GLS1 by CB839 reduced the numbers of follicular helper T (TfH) cells and activated B cells in lupus mice. The proportions of mature B cells and plasma cells were not affected. Furthermore, the hyperactivated mTOR/P70S6K/4EBP1 and NLRP3/caspase-1/IL-1β pathways in MRL/lpr mice were reversed by CB839 treatment. CONCLUSION Our study confirmed the presence of abnormal intracellular glutamine metabolism in SLE and revealed potential therapeutic targets for this disease.
Collapse
Affiliation(s)
- Xiaomei Zhang
- Department of Rheumatology and Immunology, Shengjing Hospital of China Medical University, Shenyang 110022, China
| | - Gang Wang
- Department of Neurosurgery, the First Hospital of China Medical University, Shenyang 110001, China.
| | - Ying Bi
- Department of Rheumatology and Immunology, the Fourth Hospital of China Medical University, Shenyang 110001, China
| | - Zhihang Jiang
- Department of Rheumatology and Immunology, Shengjing Hospital of China Medical University, Shenyang 110022, China
| | - Xiaofei Wang
- Department of Rheumatology and Immunology, Shengjing Hospital of China Medical University, Shenyang 110022, China
| |
Collapse
|
24
|
Nica V, Popp RA, Crișan TO, Joosten LAB. The future clinical implications of trained immunity. Expert Rev Clin Immunol 2022; 18:1125-1134. [PMID: 36062825 DOI: 10.1080/1744666x.2022.2120470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Trained Immunity (TI) refers to the long-term modulation of the innate immune response, based on previous interactions with microbes, microbial ligands or endogenous substances. Through metabolic and epigenetic reprogramming, monocytes, macrophages and neutrophils develop an enhanced capacity to mount innate immune responses to subsequent stimuli and this is persistent due to alterations at the myeloid progenitor compartment. AREAS COVERED The purpose of this article is to review the current understanding of the TI process and discuss about its potential clinical implications in the near future. We address the evidence of TI involvement in various diseases, the currently developed new therapy, and discuss how TI may lead to new clinical tools to improve existing standards of care. EXPERT OPINION The state of art in this domain has made considerable progress, linking TI-related mechanisms in multiple immune-mediated pathologies, starting with infections to autoimmune disorders and cancers. As a relatively new area of immunology, it has seen fast progress with many of its applications ready to be investigated in clinical settings.
Collapse
Affiliation(s)
- Valentin Nica
- Department of Medical Genetics, "Iuliu Hațieganu" University of Medicine and Pharmacy, Str. Pasteur nr. 6, 400349, Cluj-Napoca, Romania
| | - Radu A Popp
- Department of Medical Genetics, "Iuliu Hațieganu" University of Medicine and Pharmacy, Str. Pasteur nr. 6, 400349, Cluj-Napoca, Romania
| | - Tania O Crișan
- Department of Medical Genetics, "Iuliu Hațieganu" University of Medicine and Pharmacy, Str. Pasteur nr. 6, 400349, Cluj-Napoca, Romania
| | - Leo A B Joosten
- Department of Medical Genetics, "Iuliu Hațieganu" University of Medicine and Pharmacy, Str. Pasteur nr. 6, 400349, Cluj-Napoca, Romania.,Department of Internal Medicine and Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| |
Collapse
|
25
|
Wu J, Pang T, Lin Z, Zhao M, Jin H. The key player in the pathogenesis of environmental influence of systemic lupus erythematosus: Aryl hydrocarbon receptor. Front Immunol 2022; 13:965941. [PMID: 36110860 PMCID: PMC9468923 DOI: 10.3389/fimmu.2022.965941] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/01/2022] [Indexed: 11/28/2022] Open
Abstract
The aryl hydrocarbon receptor was previously known as an environmental receptor that modulates the cellular response to external environmental changes. In essence, the aryl hydrocarbon receptor is a cytoplasmic receptor and transcription factor that is activated by binding to the corresponding ligands, and they transmit relevant information by binding to DNA, thereby activating the transcription of various genes. Therefore, we can understand the development of certain diseases and discover new therapeutic targets by studying the regulation and function of AhR. Several autoimmune diseases, including systemic lupus erythematosus (SLE), have been connected to AhR in previous studies. SLE is a classic autoimmune disease characterized by multi-organ damage and disruption of immune tolerance. We discuss here the homeostatic regulation of AhR and its ligands among various types of immune cells, pathophysiological roles, in addition to the roles of various related cytokines and signaling pathways in the occurrence and development of SLE.
Collapse
|
26
|
Li H, Liang J, Gao Y, Liu M, Xia N, Kong W, Zheng L, Zhang Y, Li Z, Chen H, Liu S, Sun L. IGFBP2 function as a novel biomarker for active lupus nephritis. J Mol Med (Berl) 2022; 100:1479-1491. [PMID: 36008635 PMCID: PMC9470718 DOI: 10.1007/s00109-022-02241-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 07/01/2022] [Accepted: 07/25/2022] [Indexed: 11/23/2022]
Abstract
Abstract In search for new targets for the diagnosis and treatment of lupus nephritis (LN), we employed TMT-liquid chromatography-triple quadrupole mass spectrometry (TMT-LC–MS/MS) combined with RNA-seq and identified a panel of proteins that was dysregulated both at protein level and mRNA level in active LN patients compared with healthy controls. We chose to study the role of IGFBP2 since it is a relatively understudied protein in the context of LN. We further validated that IGFBP2 significantly increased and correlated with SLE activity index in active LN patients. The receiver operator characteristic (ROC) curve suggested that plasma IGFBP2 had a high diagnostic efficiency for distinguishing between inactive and active LN patients (AUC = 0.992; 95% CI = 0.974–1.000; P < 0.001). We demonstrated neutralizing IGFBP2-downregulated CD4+ T cell activation, upregulated the ratio of Treg, downregulated AKT/mTOR/4E-BP1 pathway, and significantly improved nephritis in MRL/lpr mice. In all, our work demonstrated IGFBP2 as a biomarker specific for active LN and blocking IGFBP2 could be a new target for treating LN. Key messages Plasma IGFBP2 is a promising diagnostic marker for distinguishing stable LN from active LN, and it is also a predictor for the poor prognosis of LN. Blockade of IGFBP2 can significantly improve the pathological damage of LN. IGFBP2 may regulate activation of CD4+ T and Treg ratio. Neutralizing IGFBP2 downregulates AKT/mTOR/4E-BP1 pathway.
Supplementary Information The online version contains supplementary material available at 10.1007/s00109-022-02241-z.
Collapse
Affiliation(s)
- Hui Li
- Department of Rheumatology and Immunology, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu, 210008, China.,Northern Jiangsu People's Hospital Affiliated to Clinical Medical College, Yangzhou University, Yangzhou, 225002, China
| | - Jun Liang
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, 210008, China
| | - Yingying Gao
- Department of Rheumatology and Immunology, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu, 210008, China
| | - Min Liu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Nan Xia
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, 210008, China
| | - Wei Kong
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, 210008, China
| | - Lisha Zheng
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Yikun Zhang
- PLA Strategic Support Force Characteristic Medical Center, 9 Anxiang Beili, Chaoyang District, Beijing, China
| | - Zutong Li
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, 210008, China
| | - Hongwei Chen
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, 210008, China
| | - Shanshan Liu
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, 210008, China.
| | - Lingyun Sun
- Department of Rheumatology and Immunology, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu, 210008, China. .,Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, 210008, China.
| |
Collapse
|
27
|
Robinson GA, Wilkinson MGL, Wincup C. The Role of Immunometabolism in the Pathogenesis of Systemic Lupus Erythematosus. Front Immunol 2022; 12:806560. [PMID: 35154082 PMCID: PMC8826250 DOI: 10.3389/fimmu.2021.806560] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 12/27/2021] [Indexed: 12/15/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disorder in which pathogenic abnormalities within both the innate and adaptive immune response have been described. In order to activated, proliferate and maintain this immunological response a drastic upregulation in energy metabolism is required. Recently, a greater understanding of these changes in cellular bioenergetics have provided new insight into the links between immune response and the pathogenesis of a number of diseases, ranging from cancer to diabetes and multiple sclerosis. In this review, we highlight the latest understanding of the role of immunometabolism in SLE with particular focus on the role of abnormal mitochondrial function, lipid metabolism, and mTOR signaling in the immunological phenomenon observed in the SLE. We also consider what implications this has for future therapeutic options in the management of the disease in future.
Collapse
Affiliation(s)
- George Anthony Robinson
- Department of Rheumatology, Division of Medicine, University College London, London, United Kingdom.,Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospital (UCLH) and Great Ormond Street Hospital (GOSH), University College London, London, United Kingdom
| | - Meredyth G Ll Wilkinson
- Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospital (UCLH) and Great Ormond Street Hospital (GOSH), University College London, London, United Kingdom.,Department of Rheumatology, University College London Great Ormond Street Institute of Child Health, Infection, Immunity and Inflammation Research and Teaching Department, University College London, London, United Kingdom
| | - Chris Wincup
- Department of Rheumatology, Division of Medicine, University College London, London, United Kingdom.,Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospital (UCLH) and Great Ormond Street Hospital (GOSH), University College London, London, United Kingdom
| |
Collapse
|
28
|
Schall N, Daubeuf F, Marsol C, Gizzi P, Frossard N, Bonnet D, Galzi JL, Muller S. A Selective Neutraligand for CXCL12/SDF-1α With Beneficial Regulatory Functions in MRL/Lpr Lupus Prone Mice. Front Pharmacol 2021; 12:752194. [PMID: 34744730 PMCID: PMC8566942 DOI: 10.3389/fphar.2021.752194] [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: 08/02/2021] [Accepted: 09/28/2021] [Indexed: 11/13/2022] Open
Abstract
Dysregulation of CXCL12/SDF-1-CXCR4/CD184 signaling is associated with inflammatory diseases and notably with systemic lupus erythematosus. Issued from the lead molecule chalcone-4, the first neutraligand of the CXCL12 chemokine, LIT-927 was recently described as a potent analogue with improved solubility and stability. We aimed to investigate the capacity of LIT-927 to correct immune alterations in lupus-prone MRL/lpr mice and to explore the mechanism of action implemented by this small molecule in this model. We found that in contrast to AMD3100, an antagonist of CXCR4 and agonist of CXCR7, LIT-927 reduces the excessive number of several B/T lymphocyte subsets occurring in the blood of sick MRL/lpr mice (including CD3+/CD4-/CD8-/B220+ double negative T cells). In vitro, LIT-927 downregulated the overexpression of several activation markers on splenic MRL/lpr lymphocytes. It exerted effects on the CXCR4 pathway in MRL/lpr CD4+ T spleen cells. The results underline the importance of the CXCL12/CXCR4 axis in lupus pathophysiology. They indicate that neutralizing CXCL12 by the neutraligand LIT-927 can attenuate hyperactive lymphocytes in lupus. This mode of intervention might represent a novel strategy to control a common pathophysiological mechanism occurring in inflammatory diseases.
Collapse
Affiliation(s)
- Nicolas Schall
- CNRS UMR7242, Biotechnology and Cell Signaling, Ecole Supérieure de Biotechnologie de Strasbourg, Strasbourg University/Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, France
| | - François Daubeuf
- CNRS UMR7200, Laboratoire d'innovation Thérapeutique, Faculté de Pharmacie, Strasbourg University/Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, France.,CNRS UMS3286, Plate-forme de Chimie Biologique Intégrative de Strasbourg, Strasbourg University/ Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, France
| | - Claire Marsol
- CNRS UMR7200, Laboratoire d'innovation Thérapeutique, Faculté de Pharmacie, Strasbourg University/Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, France
| | - Patrick Gizzi
- CNRS UMS3286, Plate-forme de Chimie Biologique Intégrative de Strasbourg, Strasbourg University/ Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, France
| | - Nelly Frossard
- CNRS UMR7200, Laboratoire d'innovation Thérapeutique, Faculté de Pharmacie, Strasbourg University/Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, France
| | - Dominique Bonnet
- CNRS UMR7200, Laboratoire d'innovation Thérapeutique, Faculté de Pharmacie, Strasbourg University/Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, France
| | - Jean-Luc Galzi
- CNRS UMR7242, Biotechnology and Cell Signaling, Ecole Supérieure de Biotechnologie de Strasbourg, Strasbourg University/Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, France
| | - Sylviane Muller
- CNRS UMR7242, Biotechnology and Cell Signaling, Ecole Supérieure de Biotechnologie de Strasbourg, Strasbourg University/Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, France.,Fédération Hospitalo-Universitaire (FHU) OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg University, Strasbourg, France.,University of Strasbourg Institute for Advanced Study (USIAS), Strasbourg, France
| |
Collapse
|
29
|
Thiruvengadam M, Subramanian U, Venkidasamy B, Thirupathi P, Samynathan R, Shariati MA, Rebezov M, Chung IM, Rengasamy KRR. Emerging role of nutritional short-chain fatty acids (SCFAs) against cancer via modulation of hematopoiesis. Crit Rev Food Sci Nutr 2021; 63:827-844. [PMID: 34319824 DOI: 10.1080/10408398.2021.1954874] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The understanding of gut microbiota has emerged as a significant frontier in development of strategies to maintain normal human body's homeostasis and preventing the disease development over the last decade. The composition of the gut microbiota influences the clinical benefit of immune checkpoints in patients with advanced cancer, but the mechanisms underlying this relationship are unclear. Cancer is among the leading causes of mortality worldwide. So far, there is no universal treatment for cancer and despite significant advances, a lot of improvement on cancer therapy is required. Owing to its role in preserving the host's health and maintaining cellular integrity, the human gut microbiome has recently drawn a lot of interest as a target for cancer treatment. Dietary fiber is fermented by the gut microbiota to generate short-chain fatty acids (SCFAs), such as acetate, butyrate, and propionate, which are physiologically active metabolites. SCFAs can modulate the pathophysiology of the tumor environment through various critical signaling pathways. In addition, SCFAs can bind to carcinogens and other toxic chemicals, thus facilitating their biotransformation and elimination through different excretory mechanisms. This review discusses the mechanisms of action of short-chain fatty acids in modulating hematopoiesis of various immune system cells and the resultant beneficial anti-cancer effects. It also provides future perspectives on cancer therapy.
Collapse
Affiliation(s)
- Muthu Thiruvengadam
- Department of Crop Science, College of Sanghuh Life Science, Konkuk University, Seoul, Republic of Korea
| | - Umadevi Subramanian
- Translational Research Platform for Veterinary Biologicals, Tamil Nadu Veterinary and Animal Sciences University, Chennai, India
| | - Baskar Venkidasamy
- Department of Biotechnology, Sri Shakthi Institute of Engineering and Technology, Coimbatore, India
| | - Prabhu Thirupathi
- Translational Research Platform for Veterinary Biologicals, Tamil Nadu Veterinary and Animal Sciences University, Chennai, India
| | | | - Mohammad Ali Shariati
- Department of Technology of Food Products, K.G. Razumovsky Moscow State University of Technologies and Management (The First Cossack University), Moscow, Russian Federation
| | - Maksim Rebezov
- V M Gorbatov Federal Research Center for Food Systems of Russian Academy of Sciences, Moscow, Russian Federation.,Prokhorov General Physics Institute of the Russian Academy of Science, Moscow, Russian Federation
| | - Ill-Min Chung
- Department of Crop Science, College of Sanghuh Life Science, Konkuk University, Seoul, Republic of Korea
| | - Kannan R R Rengasamy
- Green Biotechnologies Research Centre of Excellence, University of Limpopo, Mankweng, South Africa
| |
Collapse
|
30
|
Lin F, Liu Y, Tang L, Xu X, Zhang X, Song Y, Chen B, Ren Y, Yang X. Rapamycin protects against aristolochic acid nephropathy in mice by potentiating mammalian target of rapamycin‑mediated autophagy. Mol Med Rep 2021; 24:495. [PMID: 33955513 PMCID: PMC8127069 DOI: 10.3892/mmr.2021.12134] [Citation(s) in RCA: 2] [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: 01/14/2021] [Accepted: 04/04/2021] [Indexed: 12/23/2022] Open
Abstract
Autophagy serves a crucial role in the etiology of kidney diseases, including drug‑induced renal impairment, inherited kidney disease, diabetic nephropathy and aristolochic acid nephropathy (AAN) and is, therefore, a potential target for treatment. We previously demonstrated that rapamycin could attenuate AAN in mice; however, the underlying mechanism remains to be elucidated. Therefore, whether the renal protective effect of rapamycin (an autophagy activator) is related to autophagy in aristolochic acid (AA)‑treated mice was of particular interest. The pathophysiological roles of rapamycin were investigated in AA‑induced nephrotoxicity in mice and the mechanisms of rapamycin action were explored by evaluating the modulation of autophagy in rapamycin‑treated mice and cultured renal tubular epithelial cells. Supplementation with rapamycin reversed AA‑induced kidney injury in mice and improved AA‑induced autophagy damage in vivo and in vitro. Mechanistically, rapamycin inhibited the renal expression of phosphorylated (p‑)mammalian target of rapamycin (mTOR) and p‑ribosomal S6 protein kinase 1, which in turn activated renal autophagy and decreased apoptosis, probably by removing AA‑elicited damaged mitochondria and misfolded proteins. The findings of the present study demonstrated that rapamycin protects against AA‑induced nephropathy by activating the mTOR‑autophagy axis and suggested that rapamycin may be a promising pharmacological target for the treatment of AAN.
Collapse
Affiliation(s)
- Fan Lin
- Department of Nephrology, Qilu Hospital, Shandong University, Jinan, Shandong 250015, P.R. China
- Department of Nephrology, Shenzhen University General Hospital, Shenzhen, Guangdong 518055, P.R. China
| | - Yunqi Liu
- Department of Nephrology, Qilu Hospital, Shandong University, Jinan, Shandong 250015, P.R. China
- Department of Nephrology, Binzhou Medical University Hospital, Binzhou, Shandong 256600, P.R. China
| | - Lili Tang
- Clinical Laboratory, Chinese Medical Hospital of Jining, Jining, Shandong 272037, P.R. China
| | - Xiaohui Xu
- Department of Nephrology, Shenzhen University General Hospital, Shenzhen, Guangdong 518055, P.R. China
| | - Xueli Zhang
- Department of Nephrology, Shenzhen University General Hospital, Shenzhen, Guangdong 518055, P.R. China
| | - Yifan Song
- Department of Nephrology, Shenzhen University General Hospital, Shenzhen, Guangdong 518055, P.R. China
| | - Bicheng Chen
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yeping Ren
- Department of Nephrology, Shenzhen University General Hospital, Shenzhen, Guangdong 518055, P.R. China
| | - Xiangdong Yang
- Department of Nephrology, Qilu Hospital, Shandong University, Jinan, Shandong 250015, P.R. China
| |
Collapse
|
31
|
Cai B, Cai J, Yin Z, Jiang X, Yao C, Ma J, Xue Z, Miao P, Xiao Q, Cheng Y, Qin J, Guo Q, Shen N, Ye Z, Qu B, Ding H. Long non-coding RNA expression profiles in neutrophils revealed potential biomarker for prediction of renal involvement in SLE patients. Rheumatology (Oxford) 2021; 60:1734-1746. [PMID: 33068407 DOI: 10.1093/rheumatology/keaa575] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/06/2020] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE The long non-coding RNA plays an important role in inflammation and autoimmune diseases. The aim of this study is to screen and identify abnormally expressed lncRNAs in peripheral blood neutrophils of SLE patients as novel biomarkers and to explore the relationship between lncRNAs levels and clinical features, disease activity and organ damage. METHODS RNA-seq technology was used to screen differentially expressed lncRNAs in neutrophils from SLE patients and healthy donors. Based on the results of screening, candidate lncRNA levels in neutrophils of 88 SLE patients, 35 other connective disease controls, and 78 healthy controls were qualified by real-time quantitative polymerase chain reaction. RESULTS LncRNA expression profiling revealed 360 up-regulated lncRNAs and 224 down-regulated lncRNAs in neutrophils of SLE patients when compared with healthy controls. qPCR assay validated that the expression of Lnc-FOSB-1:1 was significantly decreased in neutrophils of SLE patients when compared with other CTD patients or healthy controls. It correlated negatively with SLE Disease Activity Index 2000 (SLEDAI-2K) score (r = -0.541, P < 0.001) and IFN scores (r = -0.337, P = 0.001). More importantly, decreased Lnc-FOSB-1:1 expression was associated with lupus nephritis. Lower baseline Lnc-FOSB-1:1 level was associated with higher risk of future renal involvement (within an average of 2.6 years) in patients without renal disease at baseline (P = 0.019). CONCLUSION LncRNA expression profile in neutrophils of SLE patients revealed differentially expressed lncRNAs. Validation study on Lnc-FOSB-1:1 suggest that it is a potential biomarker for prediction of near future renal involvement.
Collapse
Affiliation(s)
- Bin Cai
- Department of Rheumatology, Renji Hospital, Shanghai Institute of Rheumatology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jingyi Cai
- Department of Rheumatology, Renji Hospital, Shanghai Institute of Rheumatology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhihua Yin
- Department of Rheumatology, Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| | - Xiaoyue Jiang
- Department of Rheumatology, Renji Hospital, Shanghai Institute of Rheumatology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chao Yao
- Laboratory of Molecular Rheumatology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jianyang Ma
- China-Australia Centre for Personalized Immunology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhixin Xue
- Department of Rheumatology, Renji Hospital, Shanghai Institute of Rheumatology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ping Miao
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qingqing Xiao
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yijun Cheng
- Department of Rheumatology, Renji Hospital, Shanghai Institute of Rheumatology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jialin Qin
- Department of Rheumatology, Renji Hospital, Shanghai Institute of Rheumatology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qiang Guo
- Department of Rheumatology, Renji Hospital, Shanghai Institute of Rheumatology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Nan Shen
- Department of Rheumatology, Renji Hospital, Shanghai Institute of Rheumatology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Department of Rheumatology, Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China.,Laboratory of Molecular Rheumatology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,China-Australia Centre for Personalized Immunology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Zhizhong Ye
- Department of Rheumatology, Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| | - Bo Qu
- Department of Rheumatology, Renji Hospital, Shanghai Institute of Rheumatology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Huihua Ding
- Department of Rheumatology, Renji Hospital, Shanghai Institute of Rheumatology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
32
|
Xu R, Zhang Y, Li A, Ma Y, Cai W, Song L, Xie Y, Zhou S, Cao W, Tang X. LY‑294002 enhances the chemosensitivity of liver cancer to oxaliplatin by blocking the PI3K/AKT/HIF‑1α pathway. Mol Med Rep 2021; 24:508. [PMID: 33982772 PMCID: PMC8134878 DOI: 10.3892/mmr.2021.12147] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 04/12/2021] [Indexed: 02/07/2023] Open
Abstract
Liver cancer remains one of the leading causes of cancer deaths worldwide. The therapeutic effect of oxaliplatin on liver cancer is often limited by acquired resistance of the cancer cells. Abnormal activation of the PI3K/AKT pathway plays an important role in the acquired resistance of oxaliplatin. The present study investigated the effects of the PI3K inhibitor LY-294002 and AKT inhibitor MK2206 on the chemosensitivity of oxaliplatin-resistant liver cancer cells and the molecular mechanism involved. An oxaliplatin-resistant liver cancer cell line HepG2R was developed. MTT assay, clone formation experiments, flow cytometry and Annexin V-FITC/PI staining were used to determine the proliferation, cycle and apoptosis of HepG2R cells when oxaliplatin was combined with LY-294002 or MK2206 treatment. The effects of LY-294002 and MK-2206 on the abnormal activation of PI3K/AKT pathway and hypoxia inducible factor (HIF)-1α protein level in HepG2R cells were detected using western blotting. The results indicated that the PI3K/AKT pathway is stably activated in HepG2R cells. Compared with the AKT inhibitor MK2206, the PI3K inhibitor LY-294002 more effectively downregulated the phosphorylation levels of p85, p110α, p110β, p110γ and AKT in the PI3K/AKT pathway in HepG2R cells, and more effectively inhibited the proliferation of the cells. LY-294002 enhanced the chemotherapy sensitivity of HepG2R cells to oxaliplatin by inducing G0/G1 phase arrest and increasing the proportion of apoptotic cells. In addition, LY-294002 reduced the level of HIF-1α, which is highly expressed in HepG2R cells. It was concluded that LY-294002 enhanced the chemosensitivity of liver cancer cells to oxaliplatin by inhibiting the PI3K/AKT signaling pathway, which may be related to the inhibition of HIF-1α expression. These findings may have clinical significance for the treatment of oxaliplatin-resistant liver cancer.
Collapse
Affiliation(s)
- Ruyue Xu
- Medical School, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
| | - Yinci Zhang
- Medical School, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
| | - Amin Li
- Medical School, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
| | - Yongfang Ma
- Medical School, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
| | - Wenpeng Cai
- Medical School, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
| | - Li Song
- Medical School, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
| | - Yinghai Xie
- Institute of Environmentally Friendly Materials and Occupational Health, Anhui University of Science and Technology, Wuhu, Anhui 241000, P.R. China
| | - Shuping Zhou
- Institute of Environmentally Friendly Materials and Occupational Health, Anhui University of Science and Technology, Wuhu, Anhui 241000, P.R. China
| | - Weiya Cao
- Medical School, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
| | - Xiaolong Tang
- Medical School, Anhui University of Science and Technology, Huainan, Anhui 232001, P.R. China
| |
Collapse
|
33
|
Song X, Gao J, Liu H, Liu X, Tang K. Rapamycin alleviates renal damage in mice with systemic lupus erythematosus through improving immune response and function. Biomed Pharmacother 2021; 137:111289. [PMID: 33581650 DOI: 10.1016/j.biopha.2021.111289] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 01/08/2021] [Accepted: 01/13/2021] [Indexed: 12/13/2022] Open
Abstract
This study aimed to explore the therapeutic effect and mechanism of rapamycin (RAPA) on systemic lupus erythematosus (SLE) in BALB/C mice induced by pristane. The mice were randomly divided into 5 groups (n = 6): control, model, saline, RAPA (1 mg/kg) and RAPA (2 mg/kg). All groups were injected with pristane except control. HE staining revealed 1 mg/kg and 2 mg/kg RAPA treatments obviously alleviated pathological changes in the kidney of SLE mice such as glomeruli enlargement, hyperplasia of mesangial cells, epithelial and endothelial cells, infiltration of inflammatory cells, and edema-like degeneration of renal tubules. Compared with control group, body weights and anti-ribosomal P-protein antibody (ARPA) level of the mice in model group and saline group decreased (P < 0.05), while immune complex deposition and levels of anti-dsDNA antibody, anti-smRNP antibody and urine protein in model group and saline group increased (P < 0.05). However, compared with model group and saline group, body weights of the mice in RAPA (1 mg/kg) group and RAPA (2 mg/kg) group increased (P < 0.05), while immune complex deposition and levels of anti-dsDNA antibody, anti-smRNP antibody, ARPA, and urine protein in RAPA (1 mg/kg) group and RAPA (2 mg/kg) group decreased (P < 0.05). Compared with control group, the proportion of dentritic cells (DC) in the kidney and peripheral blood decreased while the proportion of Th1, Th2 and Th17 cells in the spleen, kidney and peripheral blood increased in model group and saline group (P < 0.05). Compared with model group and saline group, 1 mg/kg and 2 mg/kg RAPA treatments boosted the proportion of DC in the kidney and peripheral blood, reduced the proportion of Th1 and Th17 cells in the spleen, kidney and peripheral blood, and lessened the proportion of Th2 cells in the kidney and peripheral blood (P < 0.05). In conclusion, RAPA alleviated renal damage in SLE mice through improving immune response and function.
Collapse
MESH Headings
- Animals
- Antibodies, Antinuclear/blood
- Antigen-Antibody Complex/metabolism
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Disease Models, Animal
- Female
- Immunosuppressive Agents/pharmacology
- Kidney/drug effects
- Kidney/immunology
- Kidney/metabolism
- Kidney/pathology
- Lupus Erythematosus, Systemic/chemically induced
- Lupus Erythematosus, Systemic/drug therapy
- Lupus Erythematosus, Systemic/immunology
- Lupus Erythematosus, Systemic/metabolism
- Lupus Nephritis/chemically induced
- Lupus Nephritis/immunology
- Lupus Nephritis/metabolism
- Lupus Nephritis/prevention & control
- Mice, Inbred BALB C
- Sirolimus/pharmacology
- T-Lymphocytes, Helper-Inducer/drug effects
- T-Lymphocytes, Helper-Inducer/immunology
- T-Lymphocytes, Helper-Inducer/metabolism
- Terpenes
- Mice
Collapse
Affiliation(s)
- Xinghui Song
- Department of Rheumatology, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi 545027, China.
| | - Jinglin Gao
- Department of Rheumatology, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi 545027, China.
| | - Huicong Liu
- Department of Rheumatology, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi 545027, China.
| | - Xiuhua Liu
- Department of Rheumatology, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi 545027, China.
| | - Kaijiang Tang
- Department of Rheumatology, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi 545027, China.
| |
Collapse
|
34
|
Zeng Y, Hu X, Yu Z, Wang F, Zhang Z, He K, Tian H, Yu F. Immune enhancement and antioxidant effects of low molecular-weight peptides derived from Nibea japonica muscles on immune-deficient mice induced by cyclophosphamide. Process Biochem 2021. [DOI: 10.1016/j.procbio.2020.11.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
35
|
Romo-Tena J, Kaplan MJ. Immunometabolism in the pathogenesis of systemic lupus erythematosus: an update. Curr Opin Rheumatol 2020; 32:562-571. [PMID: 32826478 PMCID: PMC10463177 DOI: 10.1097/bor.0000000000000738] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW To provide an update on state-of-the-art evidence on the role of immunometabolism reprogramming in the pathogenesis of systemic lupus erythematosus (SLE). RECENT FINDINGS Mitochondrial dysfunction and enhanced oxidative stress, along with specific defects in other metabolic pathways, can promote dysregulation of innate and adaptive immune responses in SLE. These abnormalities appear to be driven by genetic and epigenetic factors, modulated by stochastic events. In addition to extensive descriptions of abnormalities in immunometabolism of lupus lymphocytes, recent studies support the critical role of dysregulation of metabolic pathways in innate immune cells including neutrophils, macrophages and dendritic cells, in SLE pathogenesis. Recent abnormalities described in lipid metabolism have been associated with SLE disease activity and related damage. Promising therapeutic strategies that target these metabolic abnormalities have recently been described in SLE. SUMMARY Fundamental new insights regarding the role of mitochondrial dysfunction in innate immune dysregulation in SLE pathogenesis have recently emerged. Defects in specific molecular pathways pertinent to immunometabolism in SLE have been described. New insights in translational medicine and promising therapeutic targets have been proposed based on these recent findings.
Collapse
Affiliation(s)
- Jorge Romo-Tena
- Systemic Autoimmunity Branch, Intramural Research Program, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Medical Science PhD Program, School of Medicine, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Mariana J. Kaplan
- Systemic Autoimmunity Branch, Intramural Research Program, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
36
|
Peng L, Wu C, Hong R, Sun Y, Qian J, Zhao J, Wang Q, Tian X, Wang Y, Li M, Zeng X. Clinical efficacy and safety of sirolimus in systemic lupus erythematosus: a real-world study and meta-analysis. Ther Adv Musculoskelet Dis 2020; 12:1759720X20953336. [PMID: 32973935 PMCID: PMC7493251 DOI: 10.1177/1759720x20953336] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 08/01/2020] [Indexed: 12/18/2022] Open
Abstract
Objective: To provide real-world data and summarize current clinical evidence on the efficacy and safety of sirolimus in active systemic lupus erythematosus (SLE) patients. Methods: This was a prospective real-world clinical study. Included SLE patients should have Systemic Lupus Erythematosus Disease Activity Index 2000 (SLEDAI-2K) ⩾ 2. They were treated with sirolimus and followed up regularly. The SLEDAI-2K, Physician Global Assessment (PGA), serological activity indices, and remission of organ manifestations were evaluated. We also performed a meta-analysis to integrate current evidence of sirolimus in SLE. Results: A total of 49 patients were included in the final analysis. After treatment, the SLEDAI-2K (6.2 ± 3.1 versus 4.0 ± 3.4, p = 0.001) decreased significantly, and the prednisone dosage was tapered successfully (9.9 ± 8.8 mg/day versus 5.9 ± 4.0 mg/day, p = 0.002). Serological activity indices also improved [complement 3 (C3): 0.690 ± 0.209 g/l versus 0.884 ± 0.219 g/l, p < 0.001; complement 4: 0.105 ± 0.059 g/l versus 0.141 ± 0.069 g/l, p < 0.001; anti-dsDNA antibody, 200 ± 178 IU/ml versus 156 ± 163 IU/ml, p = 0.022]. The remission proportions of arthritis, skin rash, and thrombocytopenia were 100%, 88.8%, and 46.2%, respectively. A total of 41.2% of lupus nephritis (LN) patients achieved renal remission, but the average 24-h urine protein level was not significantly changed. Meta-analysis enrolled five studies with 149 patients included, and revealed similar results regarding the changes of SLEDAI-2K [−3.5 (−5.0, −2.1)], C3 [0.224 (0.136, 0.311) g/l] and daily dosage of prednisone [−12.7 (−19.9, −5.6) mg/day]. Conclusion: Sirolimus might be effective and tolerated in SLE. The role of sirolimus in LN requires further study.
Collapse
Affiliation(s)
- Liying Peng
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Chanyuan Wu
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Ruping Hong
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Yiduo Sun
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Junyan Qian
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Jiuliang Zhao
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Qian Wang
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Xinping Tian
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Yanhong Wang
- Department of Epidemiology and Bio-statistics (YW), Institute of Basic Medical Science, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Mengtao Li
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, No.1 Shuai fu yuan, East City, Beijing 100730, China
| | - Xiaofeng Zeng
- Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, National Clinical Research Center for Dermatologic and Immunologic Diseases, Ministry of Science & Technology, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, No. 1 Shuaifuyuan, Beijing 100730, China
| |
Collapse
|
37
|
Maleknia S, Salehi Z, Rezaei Tabar V, Sharifi-Zarchi A, Kavousi K. An integrative Bayesian network approach to highlight key drivers in systemic lupus erythematosus. Arthritis Res Ther 2020; 22:156. [PMID: 32576231 PMCID: PMC7310461 DOI: 10.1186/s13075-020-02239-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 06/05/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND A comprehensive intuition of the systemic lupus erythematosus (SLE), as a complex and multifactorial disease, is a biological challenge. Dealing with this challenge needs employing sophisticated bioinformatics algorithms to discover the unknown aspects. This study aimed to underscore key molecular characteristics of SLE pathogenesis, which may serve as effective targets for therapeutic intervention. METHODS In the present study, the human peripheral blood mononuclear cell (PBMC) microarray datasets (n = 6), generated by three platforms, which included SLE patients (n = 220) and healthy control samples (n = 135) were collected. Across each platform, we integrated the datasets by cross-platform normalization (CPN). Subsequently, through BNrich method, the structures of Bayesian networks (BNs) were extracted from KEGG-indexed SLE, TCR, and BCR signaling pathways; the values of the node (gene) and edge (intergenic relationships) parameters were estimated within each integrated datasets. Parameters with the FDR < 0.05 were considered significant. Finally, a mixture model was performed to decipher the signaling pathway alterations in the SLE patients compared to healthy controls. RESULTS In the SLE signaling pathway, we identified the dysregulation of several nodes involved in the (1) clearance mechanism (SSB, MACROH2A2, TRIM21, H2AX, and C1Q gene family), (2) autoantigen presentation by MHCII (HLA gene family, CD80, IL10, TNF, and CD86), and (3) end-organ damage (FCGR1A, ELANE, and FCGR2A). As a remarkable finding, we demonstrated significant perturbation in CD80 and CD86 to CD28, CD40LG to CD40, C1QA and C1R to C2, and C1S to C4A edges. Moreover, we not only replicated previous studies regarding alterations of subnetworks involved in TCR and BCR signaling pathways (PI3K/AKT, MAPK, VAV gene family, AP-1 transcription factor) but also distinguished several significant edges between genes (PPP3 to NFATC gene families). Our findings unprecedentedly showed that different parameter values assign to the same node based on the pathway topology (the PIK3CB parameter values were 1.7 in TCR vs - 0.5 in BCR signaling pathway). CONCLUSIONS Applying the BNrich as a hybridized network construction method, we highlight under-appreciated systemic alterations of SLE, TCR, and BCR signaling pathways in SLE. Consequently, having such a systems biology approach opens new insights into the context of multifactorial disorders.
Collapse
Affiliation(s)
- Samaneh Maleknia
- Laboratory of Complex Biological Systems and Bioinformatics (CBB), Department of Bioinformatics, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Zahra Salehi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Vahid Rezaei Tabar
- Department of Statistics, Allameh Tabataba'i University, Tehran, Iran
- School of Biological Sciences, Institute for Research in Fundamental Sciences, Tehran, Iran
| | - Ali Sharifi-Zarchi
- Department of Computer Engineering, Sharif University of Technology, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Tehran, Iran
| | - Kaveh Kavousi
- Laboratory of Complex Biological Systems and Bioinformatics (CBB), Department of Bioinformatics, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran.
| |
Collapse
|