1
|
Lin YY, Huang CC, Ko CY, Tsai CH, Chang JW, Achudhan D, Tang CH. Omentin-1 modulates interleukin expression and macrophage polarization: Implications for rheumatoid arthritis therapy. Int Immunopharmacol 2025; 149:114205. [PMID: 39908806 DOI: 10.1016/j.intimp.2025.114205] [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: 11/24/2024] [Revised: 01/23/2025] [Accepted: 01/30/2025] [Indexed: 02/07/2025]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a systemic inflammatory and autoimmune disorder in which monocytes/macrophage infiltrate synovial membrane, differentiating into the pro- and anti-inflammatory M1 and M2 macrophage phenotypes. Omentin-1 is one of the adipokines that has anti-inflammatory and immunomodulatory effects; nevertheless, investigators have yet to elucidate the function of omentin-1 in RA development. It is still unclear how omentin-1 affects human autoimmune disease and what its beneficial role is. Thus, we show that omentin-1 exhibits a therapeutic effect on RA. METHODS Utilizing patient or animal tissue, MH7A cell-line, ELISA, and qPCR, we examined the expression of omentin-1 and inflammatory cytokines in the GEO databases. Omentin-1's effects on macrophage polarization were investigated using Immunofluorescence staining (IF) and qPCR. Additionally, the method by which omentin-1 regulates interleukins was discovered by IF labeling for STAT6 translocation, siRNA transfection, IPA software using several and pharmacological inhibitors. Omentin-1's effects were examined in an in vivo investigation using the type II collagen-induced arthritis model, micro-CT, and histological evaluation. RESULTS Results from the GSE97779 dataset and patients' tissues discovered that the level of omentin-1 and M2 macrophage markers are downregulated in human RA tissue samples compared to healthy tissue and negatively correlated with the expression of pro-inflammatory interleukins (ILs) and M1 macrophage. Stimulation of RA synovial fibroblasts with omentin-1 augmented IL-4 synthesis and subsequently enhanced anti-inflammatory ability as well as M2 polarization. The STAT6 transactivation through AMPK, PI3K, ERK, and JAK cascades regulates omentin-1-induced promotion of IL-4. Importantly, intra-articular injection of omentin-1 blocked collagen-induced arthritis-augmented pro-inflammatory response, cartilage degradation, and bone loss through upregulating IL-4 and M2 macrophages in vivo. CONCLUSION Our findings support a potential therapy goal for RA and a tenable mechanism to explain the relationship between omentin-1.
Collapse
Affiliation(s)
- Yen-You Lin
- Translational Medicine Center, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan; Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Chien-Chung Huang
- School of Medicine, China Medical University, Taichung, Taiwan; Division of Immunology and Rheumatology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Chih-Yuan Ko
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan; Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Chun-Hao Tsai
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan; Department of Sports Medicine, College of Health Care, China Medical University, Taichung, Taiwan
| | - Jun-Way Chang
- The Ph.D. Program of Biotechnology and Biomedical Industry, China Medical University, Taichung, Taiwan
| | - David Achudhan
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Chih-Hsin Tang
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan; Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan; The Ph.D. Program of Biotechnology and Biomedical Industry, China Medical University, Taichung, Taiwan; Chinese Medicine Research Center, China Medical University, Taichung, Taiwan; Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan.
| |
Collapse
|
2
|
Ma J, Li J, Chen X, Ma Y. Ojeok-san enhances platinum sensitivity in ovarian cancer by regulating adipocyte paracrine IGF1 secretion. Adipocyte 2024; 13:2282566. [PMID: 37993991 PMCID: PMC10761029 DOI: 10.1080/21623945.2023.2282566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 11/06/2023] [Indexed: 11/24/2023] Open
Abstract
BACKGROUND Platinum is a commonly used drug for ovarian cancer (OvCa) treatment, but drug resistance limits its clinical application. This study intended to delineate the effects of adipocytes on platinum resistance in OvCa. METHODS OvCa cells were maintained in the adipocyte-conditioned medium. Cell viability and apoptosis were detected by CCK-8 and flow cytometry, separately. Proliferation and apoptosis-related protein expression were assayed by western blot. The IC50 values of cisplatin and carboplatin were determined using CCK-8. IGF1 secretion and expression were assayed via ELISA and western blot, respectively. A xenograft model was established, and pathological changes were detected by H&E staining. Proliferation and apoptosis-associated protein expression was assessed via IHC. RESULTS Adipocytes promoted the viability and repressed cell apoptosis in OvCa, as well as enhancing platinum resistance, while the addition of IGF-1 R inhibitor reversed the effects of adipocytes on proliferation, apoptosis, and drug resistance of OvCa cells. Treatment with different concentrations of Ojeok-san (OJS) inhibited the adipocyte-induced platinum resistance in OvCa cells by suppressing IGF1. The combined treatment of OJS and cisplatin significantly inhibited tumour growth in vivo with good mouse tolerance. CONCLUSION In summary, OJS inhibited OvCa proliferation and platinum resistance by suppressing adipocyte paracrine IGF1 secretion.
Collapse
Affiliation(s)
- Jiong Ma
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, China
| | - Junyan Li
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, China
| | - Xuejun Chen
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, China
| | - Yanyan Ma
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, China
| |
Collapse
|
3
|
Cavazzoni A, Digiacomo G, Volta F, Alfieri R, Giovannetti E, Gnetti L, Bellini L, Galetti M, Fumarola C, Xu G, Bonelli M, La Monica S, Verzè M, Leonetti A, Eltayeb K, D'Agnelli S, Moron Dalla Tor L, Minari R, Petronini PG, Tiseo M. PD-L1 overexpression induces STAT signaling and promotes the secretion of pro-angiogenic cytokines in non-small cell lung cancer (NSCLC). Lung Cancer 2024; 187:107438. [PMID: 38100954 DOI: 10.1016/j.lungcan.2023.107438] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 11/22/2023] [Accepted: 12/07/2023] [Indexed: 12/17/2023]
Abstract
BACKGROUND Monoclonal antibodies (ICI) targeting the immune checkpoint PD-1/PD-L1 alone or in combination with chemotherapy have demonstrated relevant benefits and established new standards of care in first-line treatment for advanced non-oncogene addicted non-small cell lung cancer (NSCLC). However, a relevant percentage of NSCLC patients, even with high PD-L1 expression, did not respond to ICI, highlighting the presence of intracellular resistance mechanisms that could be dependent on high PD-L1 levels. The intracellular signaling induced by PD-L1 in tumor cells and their correlation with angiogenic signaling pathways are not yet fully elucidated. METHODS The intrinsic role of PD-L1 was initially checked in two PD-L1 overexpressing NSCLC cells by transcriptome profile and kinase array. The correlation of PD-L1 with VEGF, PECAM-1, and angiogenesis was evaluated in a cohort of advanced NSCLC patients. The secreted cytokines involved in tumor angiogenesis were assessed by Luminex assay and their effect on Huvec migration by a non-contact co-culture system. RESULTS PD-L1 overexpressing cells modulated pathways involved in tumor inflammation and JAK-STAT signaling. In NSCLC patients, PD-L1 expression was correlated with high tumor intra-vasculature. When challenged with PBMC, PD-L1 overexpressing cells produced higher levels of pro-angiogenic factors compared to parental cells, as a consequence of STAT signaling activation. This increased production of cytokines involved in tumor angiogenesis largely stimulated Huvec migration. Finally, the addition of the anti-antiangiogenic agent nintedanib significantly reduced the spread of Huvec cells when exposed to high levels of pro-angiogenic factors. CONCLUSIONS In this study, we reported that high PD-L1 modulates STAT signaling in the presence of PBMC and induces pro-angiogenic factor secretion. This could enforce the role of PD-L1 as a crucial regulator of the tumor microenvironment stimulating tumor progression, both as an inhibitor of T-cell activity and as a promoter of tumor angiogenesis.
Collapse
Affiliation(s)
- A Cavazzoni
- Department of Medicine and Surgery University of Parma, Parma, Italy.
| | - G Digiacomo
- Department of Medicine and Surgery University of Parma, Parma, Italy
| | - F Volta
- Department of Medicine and Surgery University of Parma, Parma, Italy
| | - R Alfieri
- Department of Medicine and Surgery University of Parma, Parma, Italy
| | - E Giovannetti
- Department of Medical Oncology, Amsterdam University Medical Center, VU University, Amsterdam, the Netherlands; Fondazione Pisana per la Scienza ONLUS, Pisa, Italy
| | - L Gnetti
- Pathology Unit, University Hospital of Parma, Parma, Italy
| | - L Bellini
- Italian Society of Medicine and Scientific Divulgation, SIMED, Parma, Italy
| | - M Galetti
- Department of Occupational and Environmental Medicine, Epidemiology and Hygiene, Italian Workers' Compensation Authority-INAIL, 00078 Rome, Italy
| | - C Fumarola
- Department of Medicine and Surgery University of Parma, Parma, Italy
| | - G Xu
- Department of Medical Oncology, Amsterdam University Medical Center, VU University, Amsterdam, the Netherlands
| | - M Bonelli
- Department of Medicine and Surgery University of Parma, Parma, Italy
| | - S La Monica
- Department of Medicine and Surgery University of Parma, Parma, Italy
| | - M Verzè
- Department of Medicine and Surgery University of Parma, Parma, Italy; Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - A Leonetti
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - K Eltayeb
- Department of Medicine and Surgery University of Parma, Parma, Italy
| | - S D'Agnelli
- Department of Medicine and Surgery University of Parma, Parma, Italy; Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | | | - R Minari
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - P G Petronini
- Department of Medicine and Surgery University of Parma, Parma, Italy
| | - M Tiseo
- Department of Medicine and Surgery University of Parma, Parma, Italy; Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| |
Collapse
|
4
|
Tong Y, Li X, Deng Q, Shi J, Feng Y, Bai L. Advances of the small molecule drugs regulating fibroblast-like synovial proliferation for rheumatoid arthritis. Front Pharmacol 2023; 14:1230293. [PMID: 37547337 PMCID: PMC10400780 DOI: 10.3389/fphar.2023.1230293] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 07/10/2023] [Indexed: 08/08/2023] Open
Abstract
Rheumatoid arthritis (RA) is a type of chronic autoimmune and inflammatory disease. In the pathological process of RA, the alteration of fibroblast-like synoviocyte (FLS) and its related factors is the main influence in the clinic and fundamental research. In RA, FLS exhibits a uniquely aggressive phenotype, leading to synovial hyperplasia, destruction of the cartilage and bone, and a pro-inflammatory environment in the synovial tissue for perpetuation and progression. Evidently, it is a highly promising way to target the pathological function of FLS for new anti-RA drugs. Based on this, we summed up the pathological mechanism of RA-FLS and reviewed the recent progress of small molecule drugs, including the synthetic small molecule compounds and natural products targeting RA-FLS. In the end, there were some views for further action. Compared with MAPK and NF-κB signaling pathways, the JAK/STAT signaling pathway has great potential for research as targets. A small number of synthetic small molecule compounds have entered the clinic to treat RA and are often used in combination with other drugs. Meanwhile, most natural products are currently in the experimental stage, not the clinical trial stage, such as triptolide. There is an urgent need to unremittingly develop new agents for RA.
Collapse
Affiliation(s)
- Yitong Tong
- Chengdu Second People’s Hospital, Chengdu, Sichuan, China
| | - Xinyu Li
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Qichuan Deng
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yibin Feng
- School of Chinese Medicine, The University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Lan Bai
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| |
Collapse
|
5
|
Szegvari G, Dora D, Lohinai Z. Effective Reversal of Macrophage Polarization by Inhibitory Combinations Predicted by a Boolean Protein–Protein Interaction Model. BIOLOGY 2023; 12:biology12030376. [PMID: 36979068 PMCID: PMC10045914 DOI: 10.3390/biology12030376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/17/2023] [Accepted: 02/22/2023] [Indexed: 03/03/2023]
Abstract
Background: The function and polarization of macrophages has a significant impact on the outcome of many diseases. Targeting tumor-associated macrophages (TAMs) is among the greatest challenges to solve because of the low in vitro reproducibility of the heterogeneous tumor microenvironment (TME). To create a more comprehensive model and to understand the inner workings of the macrophage and its dependence on extracellular signals driving polarization, we propose an in silico approach. Methods: A Boolean control network was built based on systematic manual curation of the scientific literature to model the early response events of macrophages by connecting extracellular signals (input) with gene transcription (output). The network consists of 106 nodes, classified as 9 input, 75 inner and 22 output nodes, that are connected by 217 edges. The direction and polarity of edges were manually verified and only included in the model if the literature plainly supported these parameters. Single or combinatory inhibitions were simulated mimicking therapeutic interventions, and output patterns were analyzed to interpret changes in polarization and cell function. Results: We show that inhibiting a single target is inadequate to modify an established polarization, and that in combination therapy, inhibiting numerous targets with individually small effects is frequently required. Our findings show the importance of JAK1, JAK3 and STAT6, and to a lesser extent STK4, Sp1 and Tyk2, in establishing an M1-like pro-inflammatory polarization, and NFAT5 in creating an anti-inflammatory M2-like phenotype. Conclusions: Here, we demonstrate a protein–protein interaction (PPI) network modeling the intracellular signalization driving macrophage polarization, offering the possibility of therapeutic repolarization and demonstrating evidence for multi-target methods.
Collapse
Affiliation(s)
- Gabor Szegvari
- Translational Medicine Institute, Semmelweis University, 1094 Budapest, Hungary
| | - David Dora
- Department of Anatomy, Histology and Embryology, Semmelweis University, 1094 Budapest, Hungary
- Correspondence: (D.D.); (Z.L.); Tel.: +36-1-2156920 (D.D.)
| | - Zoltan Lohinai
- Translational Medicine Institute, Semmelweis University, 1094 Budapest, Hungary
- Pulmonary Hospital Torokbalint, 2045 Torokbalint, Hungary
- Correspondence: (D.D.); (Z.L.); Tel.: +36-1-2156920 (D.D.)
| |
Collapse
|
6
|
Khodakarami A, Adibfar S, Karpisheh V, Abolhasani S, Jalali P, Mohammadi H, Gholizadeh Navashenaq J, Hojjat-Farsangi M, Jadidi-Niaragh F. The molecular biology and therapeutic potential of Nrf2 in leukemia. Cancer Cell Int 2022; 22:241. [PMID: 35906617 PMCID: PMC9336077 DOI: 10.1186/s12935-022-02660-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 07/19/2022] [Indexed: 02/07/2023] Open
Abstract
NF-E2-related factor 2 (Nrf2) transcription factor has contradictory roles in cancer, which can act as a tumor suppressor or a proto-oncogene in different cell conditions (depending on the cell type and the conditions of the cell environment). Nrf2 pathway regulates several cellular processes, including signaling, energy metabolism, autophagy, inflammation, redox homeostasis, and antioxidant regulation. As a result, it plays a crucial role in cell survival. Conversely, Nrf2 protects cancerous cells from apoptosis and increases proliferation, angiogenesis, and metastasis. It promotes resistance to chemotherapy and radiotherapy in various solid tumors and hematological malignancies, so we want to elucidate the role of Nrf2 in cancer and the positive point of its targeting. Also, in the past few years, many studies have shown that Nrf2 protects cancer cells, especially leukemic cells, from the effects of chemotherapeutic drugs. The present paper summarizes these studies to scrutinize whether targeting Nrf2 combined with chemotherapy would be a therapeutic approach for leukemia treatment. Also, we discussed how Nrf2 and NF-κB work together to control the cellular redox pathway. The role of these two factors in inflammation (antagonistic) and leukemia (synergistic) is also summarized.
Collapse
Affiliation(s)
- Atefeh Khodakarami
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sara Adibfar
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Vahid Karpisheh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shiva Abolhasani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pooya Jalali
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Mohammadi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | | | - Mohammad Hojjat-Farsangi
- Bioclinicum, Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden.,Department of Immunology, School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran. .,Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
7
|
Tucci G, Garufi C, Pacella I, Zagaglioni M, Pinzon Grimaldos A, Ceccarelli F, Conti F, Spinelli FR, Piconese S. Baricitinib therapy response in rheumatoid arthritis patients associates to STAT1 phosphorylation in monocytes. Front Immunol 2022; 13:932240. [PMID: 35958600 PMCID: PMC9357974 DOI: 10.3389/fimmu.2022.932240] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
Baricitinib is a Janus kinase (JAK) 1 and 2 inhibitor approved for treating rheumatoid arthritis (RA). The JAK/STAT system is essential in the intracellular signaling of different cytokines and in the activation process of the monocyte lineage. This study verifies the effects of baricitinib on STAT phosphorylation in monocytes of RA patients and evaluates the correlation between STAT phosphorylation and response to therapy. We evaluated the disease activity of patients (DAS28CRP) at baseline (T0) and after 4 and 12 weeks (T1–T3) of treatment with baricitinib, dividing them into responders (n = 7) and non-responders (n = 7) based on the reduction of DAS28CRP between T0 and T1 of at least 1.2 points. Through flow cytometry, STAT1 phosphorylation was analyzed at T0/T1/T3 in monocytes, at basal conditions and after IL2, IFNα, and IL6 stimulation. We showed that monocyte frequency decreased from T0 to T1 only in responders. Regarding the phosphorylation of STAT1, we observed a tendency for higher basal pSTAT1 in monocytes of non-responder patients and, after 4 weeks, a significant reduction of cytokine-induced pSTAT1 in monocytes of responders compared with non-responders. The single IFNα stimulation only partially recapitulated the differences in STAT1 phosphorylation between the two patient subgroups. Finally, responders showed an increased IFN signature at baseline compared with non-responders. These results may suggest that monocyte frequency and STAT1 phosphorylation in circulating monocytes could represent early markers of response to baricitinib therapy.
Collapse
Affiliation(s)
- Gloria Tucci
- Department of Internal Clinical Sciences, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Cristina Garufi
- Department of Internal Clinical Sciences, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Ilenia Pacella
- Department of Internal Clinical Sciences, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Marta Zagaglioni
- Department of Internal Clinical Sciences, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Alessandra Pinzon Grimaldos
- Department of Internal Clinical Sciences, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Fulvia Ceccarelli
- Department of Internal Clinical Sciences, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Fabrizio Conti
- Department of Internal Clinical Sciences, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Francesca Romana Spinelli
- Department of Internal Clinical Sciences, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Silvia Piconese
- Department of Internal Clinical Sciences, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
- Unità di Neuroimmunologia, IRCCS Fondazione Santa Lucia, Rome, Italy
- Laboratory Affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Rome, Italy
- *Correspondence: Silvia Piconese,
| |
Collapse
|