1
|
Aquino A, Abutalimova N, Ma Y, Ismail-zade I, Grebennik V, Rubinstein A, Kudryavtsev I, Zaikova E, Sambur D, Marichev A, Kalinina O, Bautin A, Kostareva A, Vaage J, Golovkin A. Differences in Plasma Extracellular Vesicles of Different Origin in On-Pump Versus Off-Pump Cardiac Surgery. Curr Issues Mol Biol 2024; 46:13058-13077. [PMID: 39590373 PMCID: PMC11593215 DOI: 10.3390/cimb46110779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/12/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Coronary artery bypass grafting (CABG) using cardiopulmonary bypass (CPB) causes a systemic inflammatory response that can worsen patient outcomes. Off-pump surgery has been associated with a reduced inflammatory response. The precise mechanisms and the role of extracellular vesicles (EVs) in this context are not fully understood. This study aimed to investigate the early immune response, including main T- and B-lymphocyte subsets, cytokine profiles, and plasma EVs, in patients undergoing off-pump (n = 18) and on-pump (n = 18) CABG. Thirty-six patients undergoing isolated CABG were enrolled in this randomized control study. Pre- and 24 h postoperative blood samples were analyzed for immune cell populations, cytokine levels, and plasma EV phenotyping. Off-pump CABG triggered a milder immune response than on-pump surgery. On-pump surgery led to greater changes in circulating EVs, particularly platelet- (CD62P+), endothelial- (CD31+), and B-cell-derived (CD19+), as well as platelet- and erythrocyte-derived aggregates (CD41+CD235a+). Levels of platelet-derived EVs, expressing both constitutional and activation markers (CD41+CD62P+) decreased in both groups of patients 24 h after surgery. On-pump cardiac procedures led to an increase in T-regulatory cell-derived EVs (CD73+CD39+), suggesting a potential mechanism for immune suppression compared to off-pump surgery. There were numerous correlations between EV levels and cytokine profiles following on-pump surgery, hinting at a close relationship. Leucocyte-derived EVs exhibited positive correlations with each other and with GRO but showed negative correlations with endothelial-derived EVs (CD90+ and CD31+). Additionally, CD73+ EVs demonstrated positive correlations with platelet counts and with erythrocyte-derived CD235a+ EVs. EV changes were significantly greater after on-pump surgery, highlighting a more pronounced response to this type of surgery and emphasizing the role of EVs as regulators of post-surgical inflammation.
Collapse
Affiliation(s)
- Arthur Aquino
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.A.); (N.A.); (Y.M.); (I.I.-z.); (V.G.); (A.R.); (I.K.); (E.Z.); (D.S.); (A.M.); (O.K.); (A.B.); (A.K.)
| | - Napisat Abutalimova
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.A.); (N.A.); (Y.M.); (I.I.-z.); (V.G.); (A.R.); (I.K.); (E.Z.); (D.S.); (A.M.); (O.K.); (A.B.); (A.K.)
| | - Yi Ma
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.A.); (N.A.); (Y.M.); (I.I.-z.); (V.G.); (A.R.); (I.K.); (E.Z.); (D.S.); (A.M.); (O.K.); (A.B.); (A.K.)
| | - Imran Ismail-zade
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.A.); (N.A.); (Y.M.); (I.I.-z.); (V.G.); (A.R.); (I.K.); (E.Z.); (D.S.); (A.M.); (O.K.); (A.B.); (A.K.)
| | - Vadim Grebennik
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.A.); (N.A.); (Y.M.); (I.I.-z.); (V.G.); (A.R.); (I.K.); (E.Z.); (D.S.); (A.M.); (O.K.); (A.B.); (A.K.)
| | - Artem Rubinstein
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.A.); (N.A.); (Y.M.); (I.I.-z.); (V.G.); (A.R.); (I.K.); (E.Z.); (D.S.); (A.M.); (O.K.); (A.B.); (A.K.)
- Institute of Experimental Medicine, 197022 St. Petersburg, Russia
| | - Igor Kudryavtsev
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.A.); (N.A.); (Y.M.); (I.I.-z.); (V.G.); (A.R.); (I.K.); (E.Z.); (D.S.); (A.M.); (O.K.); (A.B.); (A.K.)
- Institute of Experimental Medicine, 197022 St. Petersburg, Russia
| | - Ekatherina Zaikova
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.A.); (N.A.); (Y.M.); (I.I.-z.); (V.G.); (A.R.); (I.K.); (E.Z.); (D.S.); (A.M.); (O.K.); (A.B.); (A.K.)
| | - Darina Sambur
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.A.); (N.A.); (Y.M.); (I.I.-z.); (V.G.); (A.R.); (I.K.); (E.Z.); (D.S.); (A.M.); (O.K.); (A.B.); (A.K.)
| | - Alexander Marichev
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.A.); (N.A.); (Y.M.); (I.I.-z.); (V.G.); (A.R.); (I.K.); (E.Z.); (D.S.); (A.M.); (O.K.); (A.B.); (A.K.)
| | - Olga Kalinina
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.A.); (N.A.); (Y.M.); (I.I.-z.); (V.G.); (A.R.); (I.K.); (E.Z.); (D.S.); (A.M.); (O.K.); (A.B.); (A.K.)
| | - Andrey Bautin
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.A.); (N.A.); (Y.M.); (I.I.-z.); (V.G.); (A.R.); (I.K.); (E.Z.); (D.S.); (A.M.); (O.K.); (A.B.); (A.K.)
| | - Anna Kostareva
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.A.); (N.A.); (Y.M.); (I.I.-z.); (V.G.); (A.R.); (I.K.); (E.Z.); (D.S.); (A.M.); (O.K.); (A.B.); (A.K.)
| | - Jarle Vaage
- Oslo University Hospital, University of Oslo, 0372 Oslo, Norway;
| | - Alexey Golovkin
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.A.); (N.A.); (Y.M.); (I.I.-z.); (V.G.); (A.R.); (I.K.); (E.Z.); (D.S.); (A.M.); (O.K.); (A.B.); (A.K.)
| |
Collapse
|
2
|
Poznyak AV, Yakovlev AA, Popov MА, Zhigmitova EB, Sukhorukov VN, Orekhov AN. Atherosclerosis originating from childhood: Specific features. J Biomed Res 2024; 38:233-240. [PMID: 38777340 PMCID: PMC11144930 DOI: 10.7555/jbr.37.20230198] [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: 08/21/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 05/25/2024] Open
Abstract
Atherosclerosis is extremely widespread. Traditionally, it is considered a disease of older people, who most often experience problems with the heart and blood vessels. While much attention from the scientific community has been paid to studying the association between aging and atherosclerosis, as well as its consequences, there is evidence that atherosclerosis occurs at an early age. Atherosclerosis may form both during intrauterine development and in childhood. Nutrition plays an important role in childhood atherosclerosis, along with previous infectious diseases and excess weight of both the child and the mother. In the present review, we examined the development of atherosclerosis and the prerequisites in childhood.
Collapse
Affiliation(s)
| | - Alexey A. Yakovlev
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 109240, Russia
| | - Mikhail А. Popov
- Department of Cardiac Surgery, Moscow Regional Research and Clinical Institute, Moscow 129110, Russia
| | - Elena B. Zhigmitova
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow 125315, Russia
| | - Vasily N. Sukhorukov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow 125315, Russia
| | - Alexander N. Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow 125315, Russia
| |
Collapse
|
3
|
Li XQ, Yamazaki T, He T, Alam MM, Liu J, Trivett AL, Sveinbjørnsson B, Rekdal Ø, Galluzzi L, Oppenheim JJ, Yang D. LTX-315 triggers anticancer immunity by inducing MyD88-dependent maturation of dendritic cells. Front Immunol 2024; 15:1332922. [PMID: 38545099 PMCID: PMC10967226 DOI: 10.3389/fimmu.2024.1332922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 02/22/2024] [Indexed: 04/10/2024] Open
Abstract
LTX-315 is a synthetic cationic oncolytic peptide with potent anticancer activity but limited toxicity for non-malignant cells. LTX-315 induces both immunogenic tumor cell death and generation of tumor-specific immune responses in multiple experimental tumor models. Given the central role of dendritic cell (DC) maturation in the induction of antigen-specific immunity, we investigated the effect of LTX-315 treatment on the maturation of tumor-infiltrating DCs (TiDCs) and the generation of anti-melanoma immunity. We found that LTX-315 treatment induces the maturation of DCs, both indirectly through the release of cancer cell-derived damage-associated molecular patterns (DAMPs)/alarmins and nucleic acids (DNA and RNA) capable of triggering distinct Toll-like receptor (TLR) signaling, and, directly by activating TLR7. The latter results in the ignition of multiple intracellular signaling pathways that promotes DC maturation, including NF-κB, mitogen activated protein kinases (MAPKs), and inflammasome signaling, as well as increased type 1 interferon production. Critically, the effects of LTX-315 on DCs the consequent promotion of anti-melanoma immunity depend on the cytosolic signal transducer myeloid differentiation response gene 88 (MyD88). These results cast light on the mechanisms by which LTX-315 induces DC maturation and hence elicits anticancer immunity, with important implications for the use of LTX-315 as an anticancer immunotherapeutic.
Collapse
Affiliation(s)
- Xiao-Qing Li
- Department of Biochemistry and Molecular Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin Medical University, Tianjin, China
- Laboratory of Cancer Innovation, Frederick National Laboratory for Cancer Research, Center for Cancer Research, National Cancer Institute, Frederick, MD, United States
| | - Takahiro Yamazaki
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States
| | - Tianzhen He
- Laboratory of Cancer Innovation, Frederick National Laboratory for Cancer Research, Center for Cancer Research, National Cancer Institute, Frederick, MD, United States
| | - Md Masud Alam
- Laboratory of Cancer Innovation, Frederick National Laboratory for Cancer Research, Center for Cancer Research, National Cancer Institute, Frederick, MD, United States
| | - Jia Liu
- Laboratory of Cancer Innovation, Frederick National Laboratory for Cancer Research, Center for Cancer Research, National Cancer Institute, Frederick, MD, United States
| | - Anna L. Trivett
- Laboratory of Cancer Innovation, Frederick National Laboratory for Cancer Research, Center for Cancer Research, National Cancer Institute, Frederick, MD, United States
| | | | | | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States
- Sandra and Edward Meyer Cancer Center, New York, NY, United States
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, United States
| | - Joost J. Oppenheim
- Laboratory of Cancer Innovation, Frederick National Laboratory for Cancer Research, Center for Cancer Research, National Cancer Institute, Frederick, MD, United States
| | - De Yang
- Laboratory of Cancer Innovation, Frederick National Laboratory for Cancer Research, Center for Cancer Research, National Cancer Institute, Frederick, MD, United States
| |
Collapse
|
4
|
Yoo HJ, Yi Y, Kang Y, Kim SJ, Yoon YI, Tran PH, Kang T, Kim MK, Han J, Tak E, Ahn CS, Song GW, Park GC, Lee SG, Kim JJ, Jung DH, Hwang S, Kim N. Reduced Ceramides Are Associated with Acute Rejection in Liver Transplant Patients and Skin Graft and Hepatocyte Transplant Mice, Reducing Tolerogenic Dendritic Cells. Mol Cells 2023; 46:688-699. [PMID: 37968983 PMCID: PMC10654454 DOI: 10.14348/molcells.2023.0104] [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: 06/29/2023] [Revised: 08/11/2023] [Accepted: 08/22/2023] [Indexed: 11/17/2023] Open
Abstract
We set up this study to understand the underlying mechanisms of reduced ceramides on immune cells in acute rejection (AR). The concentrations of ceramides and sphingomyelins were measured in the sera from hepatic transplant patients, skin graft mice and hepatocyte transplant mice by liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS). Serum concentrations of C24 ceramide, C24:1 ceramide, C16:0 sphingomyelin, and C18:1 sphingomyelin were lower in liver transplantation (LT) recipients with than without AR. Comparisons with the results of LT patients with infection and cardiac transplant patients with cardiac allograft vasculopathy in humans and in mouse skin graft and hepatocyte transplant models suggested that the reduced C24 and C24:1 ceramides were specifically involved in AR. A ceramide synthase inhibitor, fumonisin B1 exacerbated allogeneic immune responses in vitro and in vivo, and reduced tolerogenic dendritic cells (tDCs), while increased P3-like plasmacytoid DCs (pDCs) in the draining lymph nodes from allogeneic skin graft mice. The results of mixed lymphocyte reactions with ceranib-2, an inhibitor of ceramidase, and C24 ceramide also support that increasing ceramide concentrations could benefit transplant recipients with AR. The results suggest increasing ceramides as novel therapeutic target for AR, where reduced ceramides were associated with the changes in DC subsets, in particular tDCs.
Collapse
Affiliation(s)
- Hyun Ju Yoo
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
- Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
- Convergence Medicine Research Center, Asan Medical Center, Seoul 05505, Korea
- Digestive Disease Research Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Yeogyeong Yi
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
- Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Yoorha Kang
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
- Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Su Jung Kim
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
- Convergence Medicine Research Center, Asan Medical Center, Seoul 05505, Korea
| | - Young-In Yoon
- Division of Liver Transplantation and Hepatobiliary Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Phuc Huu Tran
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
- Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Taewook Kang
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Min Kyung Kim
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
- Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Jaeseok Han
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
- Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Eunyoung Tak
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
- Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Chul-Soo Ahn
- Division of Liver Transplantation and Hepatobiliary Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Gi-Won Song
- Division of Liver Transplantation and Hepatobiliary Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Gil-Chun Park
- Division of Liver Transplantation and Hepatobiliary Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Sung-Gyu Lee
- Division of Liver Transplantation and Hepatobiliary Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Jae-Joong Kim
- Division of Cardiology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Dong-Hwan Jung
- Division of Liver Transplantation and Hepatobiliary Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Shin Hwang
- Division of Liver Transplantation and Hepatobiliary Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Nayoung Kim
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
- Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| |
Collapse
|
5
|
Martins S, António N, Rodrigues R, Carvalheiro T, Tomaz C, Gonçalves L, Paiva A. Role of monocytes and dendritic cells in cardiac reverse remodelling after cardiac resynchronization therapy. BMC Cardiovasc Disord 2023; 23:558. [PMID: 37968611 PMCID: PMC10652525 DOI: 10.1186/s12872-023-03574-4] [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: 07/21/2023] [Accepted: 10/22/2023] [Indexed: 11/17/2023] Open
Abstract
BACKGROUND AND AIMS Monocytes and dendritic cells (DC) are both key inflammatory cells, with recognized effects on cardiac repair. However, there are distinct subsets of monocytes with potential for beneficial or detrimental effects on heart failure (HF) pathogenesis. The connection between reverse cardiac remodelling, the potential anti-inflammatory effect of cardiac resynchronization therapy (CRT) and monocytes and DC homeostasis in HF is far from being understood. We hypothesized that monocytes and DC play an important role in cardiac reverse remodelling and CRT response. Therefore, we aimed to assess the potential role of baseline peripheral levels of blood monocytes and DC subsets and their phenotypic and functional activity for CRT response, in HF patients. As a secondary objective, we aimed to evaluate the impact of CRT on peripheral blood monocytes and DC subsets, by comparing baseline and post CRT circulating levels and phenotypic and functional activity. METHODS Forty-one patients with advanced HF scheduled for CRT were included in this study. The quantification and phenotypic determination of classical (cMo), intermediate (iMo) and non-classical monocytes (ncMo), as well as of myeloid (mDC) and plasmacytoid DC (pDC) were performed by flow cytometry in a FACSCanto™II (BD) flow cytometer. The functional characterization of total monocytes and mDC was performed by flow cytometry in a FACSCalibur flow cytometer, after in vitro stimulation with lipopolysaccharide from Escherichia coli plus interferon (IFN)-γ, in the presence of Brefeldina A. Comparisons between the control and the patient group, and between responders and non-responders to CRT were performed. RESULTS Compared to the control group, HF population presented a significantly lower frequency of pDC at baseline and a higher proportion of monocytes and mDC producing IL-6 and IL-1β, both before and 6-months after CRT (T6). There was a remarkable decrease of cMo and an increase of iMo after CRT, only in responders. The responder group also presented higher ncMo values at T6 compared to the non-responder group. Both responders and non-responders presented a decrease in the expression of CD86 in all monocyte and DC populations after CRT. Moreover, in non-responders, the increased frequency of IL-6-producing DC persisted after CRT. CONCLUSION Our study provides new knowledge about the possible contribution of pDC and monocytes subsets to cardiac reverse remodelling and response to CRT. Additionally, CRT is associated with a reduction on CD86 expression by monocytes and DC subsets and in their potential to produce pro-inflammatory cytokines, contributing, at least in part, for the well described anti-inflammatory effects of CRT in HF patients.
Collapse
Affiliation(s)
- Sílvia Martins
- Health Sciences Research Centre, University of Beira Interior (CICS-UBI), 6200-506, Covilhã, Portugal
- Instituto Politécnico de Castelo Branco, ESALD-Dr. Lopes Dias Health School, Ciências Biomédicas Laboratoriais, Castelo Branco, Portugal
- Department of Clinical Pathology, Centro Hospitalar Universitário Cova da Beira, Quinta Do Alvito, 6200-251, Covilhã, Portugal
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
| | - Natália António
- Cardiology Department, Centro Hospitalar E Universitário de Coimbra, Coimbra, Portugal
- Institute of Pharmacology and Experimental Therapeutics/iCBR, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Ricardo Rodrigues
- Department of Clinical Pathology, Centro Hospitalar Universitário Cova da Beira, Quinta Do Alvito, 6200-251, Covilhã, Portugal
| | - Tiago Carvalheiro
- Centro Do Sangue E da Transplantação de Coimbra, Instituto Português Do Sangue E da Transplantação, Coimbra, Portugal
| | - Cândida Tomaz
- Health Sciences Research Centre, University of Beira Interior (CICS-UBI), 6200-506, Covilhã, Portugal
- Chemistry Department, University of Beira Interior, Covilhã, Portugal
| | - Lino Gonçalves
- Cardiology Department, Centro Hospitalar E Universitário de Coimbra, Coimbra, Portugal
- Institute of Pharmacology and Experimental Therapeutics/iCBR, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Artur Paiva
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal.
- Department of Clinical Pathology, Flow Cytometry Unit, Centro Hospitalar E Universitário de Coimbra, Coimbra, Portugal.
- Instituto Politécnico de Coimbra, ESTESC-Coimbra Health School, Ciências Biomédicas Laboratoriais, Coimbra, Portugal.
- Unidade Funcional de Citometria de Fluxo, Centro Hospitalar E Universitário de Coimbra, Praceta Mota Pinto, 3000-075, Coimbra, Portugal.
| |
Collapse
|
6
|
Márquez-Sánchez AC, Koltsova EK. Immune and inflammatory mechanisms of abdominal aortic aneurysm. Front Immunol 2022; 13:989933. [PMID: 36275758 PMCID: PMC9583679 DOI: 10.3389/fimmu.2022.989933] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is a life-threatening cardiovascular disease. Immune-mediated infiltration and a destruction of the aortic wall during AAA development plays significant role in the pathogenesis of this disease. While various immune cells had been found in AAA, the mechanisms of their activation and function are still far from being understood. A better understanding of mechanisms regulating the development of aberrant immune cell activation in AAA is essential for the development of novel preventive and therapeutic approaches. In this review we summarize current knowledge about the role of immune cells in AAA and discuss how pathogenic immune cell activation is regulated in this disease.
Collapse
|
7
|
Mulherkar TH, Gómez DJ, Sandel G, Jain P. Co-Infection and Cancer: Host–Pathogen Interaction between Dendritic Cells and HIV-1, HTLV-1, and Other Oncogenic Viruses. Viruses 2022; 14:v14092037. [PMID: 36146843 PMCID: PMC9503663 DOI: 10.3390/v14092037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/08/2022] [Accepted: 09/12/2022] [Indexed: 11/16/2022] Open
Abstract
Dendritic cells (DCs) function as a link between innate and adaptive immune responses. Retroviruses HIV-1 and HTLV-1 modulate DCs to their advantage and utilize them to propagate infection. Coinfection of HTLV-1 and HIV-1 has implications for cancer malignancies. Both viruses initially infect DCs and propagate the infection to CD4+ T cells through cell-to-cell transmission using mechanisms including the formation of virologic synapses, viral biofilms, and conduits. These retroviruses are both neurotrophic with neurovirulence determinants. The neuropathogenesis of HIV-1 and HTLV-1 results in neurodegenerative diseases such as HIV-associated neurocognitive disorders (HAND) and HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). Infected DCs are known to traffic to the brain (CNS) and periphery (PNS, lymphatics) to induce neurodegeneration in HAND and HAM/TSP patients. Elevated levels of neuroinflammation have been correlated with cognitive decline and impairment of motor control performance. Current vaccinations and therapeutics for HIV-1 and HTLV-1 are assessed and can be applied to patients with HIV-1-associated cancers and adult T cell leukemia/lymphoma (ATL). These diseases caused by co-infections can result in both neurodegeneration and cancer. There are associations with cancer malignancies and HIV-1 and HTLV-1 as well as other human oncogenic viruses (EBV, HBV, HCV, HDV, and HPV). This review contains current knowledge on DC sensing of HIV-1 and HTLV-1 including DC-SIGN, Tat, Tax, and current viral therapies. An overview of DC interaction with oncogenic viruses including EBV, Hepatitis viruses, and HPV is also provided. Vaccines and therapeutics targeting host–pathogen interactions can provide a solution to co-infections, neurodegeneration, and cancer.
Collapse
Affiliation(s)
- Tania H. Mulherkar
- Department of Microbiology and Immunology, Drexel University, College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | - Daniel Joseph Gómez
- Department of Microbiology and Immunology, Drexel University, College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
- Department of Biological Sciences, California State University, 25800 Carlos Bee Blvd, Hayward, CA 94542, USA
| | - Grace Sandel
- Department of Microbiology and Immunology, Drexel University, College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | - Pooja Jain
- Department of Microbiology and Immunology, Drexel University, College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
- Correspondence:
| |
Collapse
|
8
|
From Diabetes to Atherosclerosis: Potential of Metformin for Management of Cardiovascular Disease. Int J Mol Sci 2022; 23:ijms23179738. [PMID: 36077136 PMCID: PMC9456496 DOI: 10.3390/ijms23179738] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/20/2022] [Accepted: 08/22/2022] [Indexed: 11/17/2022] Open
Abstract
Atherosclerosis is a common cause of cardiovascular disease, which, in turn, is often fatal. Today, we know a lot about the pathogenesis of atherosclerosis. However, the main knowledge is that the disease is extremely complicated. The development of atherosclerosis is associated with more than one molecular mechanism, each making a significant contribution. These mechanisms include endothelial dysfunction, inflammation, mitochondrial dysfunction, oxidative stress, and lipid metabolism disorders. This complexity inevitably leads to difficulties in treatment and prevention. One of the possible therapeutic options for atherosclerosis and its consequences may be metformin, which has already proven itself in the treatment of diabetes. Both diabetes and atherosclerosis are complex metabolic diseases, the pathogenesis of which involves many different mechanisms, including those common to both diseases. This makes metformin a suitable candidate for investigating its efficacy in cardiovascular disease. In this review, we highlight aspects such as the mechanisms of action and targets of metformin, in addition to summarizing the available data from clinical trials on the effective reduction of cardiovascular risks.
Collapse
|
9
|
Poznyak AV, Sadykhov NK, Kartuesov AG, Borisov EE, Sukhorukov VN, Orekhov AN. Atherosclerosis Specific Features in Chronic Kidney Disease (CKD). Biomedicines 2022; 10:biomedicines10092094. [PMID: 36140195 PMCID: PMC9495595 DOI: 10.3390/biomedicines10092094] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/20/2022] [Accepted: 08/22/2022] [Indexed: 11/25/2022] Open
Abstract
Atherosclerosis is the major cause of cardiovascular disease, leading to a high mortality rate worldwide. Several risk factors are known to favor atherogenesis, among which are high blood pressure, smoking, diabetes mellitus, and others. Chronic kidney disease is another serious health problem associated with significant health care costs, morbidity, and mortality. Chronic kidney disease shares several risk factors with atherosclerosis and cardiovascular diseases, such as hypertension and diabetes mellitus. Additional risk factors for cardiovascular disease development should be considered in patients with chronic kidney disease. Interestingly, patients suffering from chronic kidney disease are more prone to cardiovascular problems than the general population. Moreover, chronic kidney disease is characterized by an increased atherosclerotic burden from the very early stages. The purpose of this review was to summarize data on atherosclerosis in chronic kidney disease, highlighting the specific features of the disease combination.
Collapse
Affiliation(s)
- Anastasia V. Poznyak
- Institute for Atherosclerosis Research, Osennyaya 4-1-207, 121609 Moscow, Russia
- Correspondence: (A.V.P.); (A.N.O.)
| | - Nikolay K. Sadykhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315 Moscow, Russia
| | - Andrey G. Kartuesov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315 Moscow, Russia
| | - Evgeny E. Borisov
- Petrovsky National Research Centre of Surgery, Abrikosovsky Lane, 119991 Moscow, Russia
| | - Vasily N. Sukhorukov
- Institute for Atherosclerosis Research, Osennyaya 4-1-207, 121609 Moscow, Russia
- Petrovsky National Research Centre of Surgery, Abrikosovsky Lane, 119991 Moscow, Russia
| | - Alexander N. Orekhov
- Institute for Atherosclerosis Research, Osennyaya 4-1-207, 121609 Moscow, Russia
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315 Moscow, Russia
- Petrovsky National Research Centre of Surgery, Abrikosovsky Lane, 119991 Moscow, Russia
- Correspondence: (A.V.P.); (A.N.O.)
| |
Collapse
|
10
|
Poznyak AV, Sadykhov NK, Kartuesov AG, Borisov EE, Melnichenko AA, Grechko AV, Orekhov AN. Hypertension as a risk factor for atherosclerosis: Cardiovascular risk assessment. Front Cardiovasc Med 2022; 9:959285. [PMID: 36072873 PMCID: PMC9441708 DOI: 10.3389/fcvm.2022.959285] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 07/18/2022] [Indexed: 11/29/2022] Open
Abstract
Atherosclerosis is a predecessor of numerous cardiovascular diseases (CVD), which often lead to morbidity and mortality. Despite the knowledge of the pathogenesis of atherosclerosis, an essential gap in our understanding is the exact trigger mechanism. A wide range of risk factors have been discovered; however, a majority of them are too general to clarify the launching mechanism of atherogenesis. Some risk factors are permanent (age, gender, genetic heritage) and others can be modified [tobacco smoking, physical inactivity, poor nutrition, high blood pressure, type 2 diabetes (T2D), dyslipidemia, and obesity]. All of them have to be taken into account. In the scope of this review, our attention is focused on hypertension, which is considered the most widespread among all modifiable risk factors for atherosclerosis development. Moreover, high blood pressure is the most investigated risk factor. The purpose of this review is to summarize the data on hypertension as a risk factor for atherosclerosis development and the risk assessment.
Collapse
Affiliation(s)
- Anastasia V. Poznyak
- Institute for Atherosclerosis Research, Moscow, Russia
- *Correspondence: Anastasia V. Poznyak,
| | | | - Andrey G. Kartuesov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia
| | | | - Alexandra A. Melnichenko
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Andrey V. Grechko
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow, Russia
| | - Alexander N. Orekhov
- Institute for Atherosclerosis Research, Moscow, Russia
- Petrovsky National Research Centre of Surgery, Moscow, Russia
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia
| |
Collapse
|
11
|
Bellini R, Bonacina F, Norata GD. Crosstalk between dendritic cells and T lymphocytes during atherogenesis: Focus on antigen presentation and break of tolerance. Front Cardiovasc Med 2022; 9:934314. [PMID: 35966516 PMCID: PMC9365967 DOI: 10.3389/fcvm.2022.934314] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 07/05/2022] [Indexed: 12/14/2022] Open
Abstract
Atherosclerosis is a chronic disease resulting from an impaired lipid and immune homeostasis, where the interaction between innate and adaptive immune cells leads to the promotion of atherosclerosis-associated immune-inflammatory response. Emerging evidence has suggested that this response presents similarities to the reactivity of effector immune cells toward self-epitopes, often as a consequence of a break of tolerance. In this context, dendritic cells, a heterogeneous population of antigen presenting cells, play a key role in instructing effector T cells to react against foreign antigens and T regulatory cells to maintain tolerance against self-antigens and/or to patrol for self-reactive effector T cells. Alterations in this delicate balance appears to contribute to atherogenesis. The aim of this review is to discuss different DC subsets, and their role in atherosclerosis as well as in T cell polarization. Moreover, we will discuss how loss of T cell tolerogenic phenotype participates to the immune-inflammatory response associated to atherosclerosis and how a better understanding of these mechanisms might result in designing immunomodulatory therapies targeting DC-T cell crosstalk for the treatment of atherosclerosis-related inflammation.
Collapse
Affiliation(s)
- Rossella Bellini
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Fabrizia Bonacina
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
- *Correspondence: Fabrizia Bonacina,
| | - Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
- Center for the Study of Atherosclerosis, E. Bassini Hospital, Cinisello Balsamo, Milan, Italy
- Giuseppe Danilo Norata,
| |
Collapse
|
12
|
Blagov AV, Grechko AV, Nikiforov NG, Zhuravlev AD, Sadykhov NK, Orekhov AN. Effects of Metabolic Disorders in Immune Cells and Synoviocytes on the Development of Rheumatoid Arthritis. Metabolites 2022; 12:metabo12070634. [PMID: 35888759 PMCID: PMC9324614 DOI: 10.3390/metabo12070634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 06/30/2022] [Accepted: 07/07/2022] [Indexed: 02/05/2023] Open
Abstract
Rheumatoid arthritis (RA) is a progressive autoimmune disease that affects the joints. It has been proven that, with the development of RA, there are changes in the metabolism of cells located in the focus of inflammation. In this article, we describe the connection between metabolism and inflammation in the context of rheumatoid arthritis. We consider in detail the changes in metabolic processes and their subsequent immunomodulatory effects. In particular, we consider how changes in mitochondrial functioning lead to the modulation of metabolism in rheumatoid arthritis. We also describe the main features of the metabolism in cells present in the synovial membrane during inflammation, and we discuss possible targets for the therapy of rheumatoid arthritis.
Collapse
Affiliation(s)
- Alexander V. Blagov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 8 Baltiiskaya Street, 125315 Moscow, Russia; (N.G.N.); (N.K.S.)
- Correspondence: (A.V.B.); (A.N.O.)
| | - Andrey V. Grechko
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 14–3 Solyanka Street, 109240 Moscow, Russia;
| | - Nikita G. Nikiforov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 8 Baltiiskaya Street, 125315 Moscow, Russia; (N.G.N.); (N.K.S.)
| | - Alexander D. Zhuravlev
- Petrovsky National Research Centre of Surgery, AP Avtsyn Institute of Human Morphology, 117418 Moscow, Russia;
| | - Nikolay K. Sadykhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 8 Baltiiskaya Street, 125315 Moscow, Russia; (N.G.N.); (N.K.S.)
| | - Alexander N. Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 8 Baltiiskaya Street, 125315 Moscow, Russia; (N.G.N.); (N.K.S.)
- Correspondence: (A.V.B.); (A.N.O.)
| |
Collapse
|
13
|
Blagov AV, Grechko AV, Nikiforov NG, Borisov EE, Sadykhov NK, Orekhov AN. Role of Impaired Mitochondrial Dynamics Processes in the Pathogenesis of Alzheimer’s Disease. Int J Mol Sci 2022; 23:ijms23136954. [PMID: 35805958 PMCID: PMC9266759 DOI: 10.3390/ijms23136954] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/17/2022] [Accepted: 06/21/2022] [Indexed: 02/06/2023] Open
Abstract
Mitochondrial dysfunction is now recognized as a contributing factor to neurodegenerative diseases, including Alzheimer’s disease (AD). Mitochondria are signaling organelles with a variety of functions ranging from energy production to the regulation of cellular metabolism, energy homeostasis, and response to stress. The successful functioning of these complex processes is critically dependent on the accuracy of mitochondrial dynamics, which includes the ability of mitochondria to change shape and position in the cell, which is necessary to maintain proper function and quality control, especially in polarized cells such as neurons. There has been much evidence to suggest that the disruption of mitochondrial dynamics may play a critical role in the pathogenesis of AD. This review highlights aspects of altered mitochondrial dynamics in AD that may contribute to the etiology of this debilitating condition. We also discuss therapeutic strategies to improve mitochondrial dynamics and function that may provide an alternative treatment approach.
Collapse
Affiliation(s)
- Alexander V. Blagov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 8 Baltiiskaya Street, 125315 Moscow, Russia; (N.G.N.); (N.K.S.)
- Correspondence: (A.V.B.); (A.N.O.)
| | - Andrey V. Grechko
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 14-3 Solyanka Street, 109240 Moscow, Russia;
| | - Nikita G. Nikiforov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 8 Baltiiskaya Street, 125315 Moscow, Russia; (N.G.N.); (N.K.S.)
| | - Evgeny E. Borisov
- Petrovsky National Research Centre of Surgery, AP Avtsyn Institute of Human Morphology, 117418 Moscow, Russia;
| | - Nikolay K. Sadykhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 8 Baltiiskaya Street, 125315 Moscow, Russia; (N.G.N.); (N.K.S.)
| | - Alexander N. Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 8 Baltiiskaya Street, 125315 Moscow, Russia; (N.G.N.); (N.K.S.)
- Correspondence: (A.V.B.); (A.N.O.)
| |
Collapse
|
14
|
Vieira VA, Herbert N, Cromhout G, Adland E, Goulder P. Role of Early Life Cytotoxic T Lymphocyte and Natural Killer Cell Immunity in Paediatric HIV Cure/Remission in the Anti-Retroviral Therapy Era. Front Immunol 2022; 13:886562. [PMID: 35634290 PMCID: PMC9130627 DOI: 10.3389/fimmu.2022.886562] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/13/2022] [Indexed: 11/13/2022] Open
Abstract
Only three well-characterised cases of functional cure have been described in paediatric HIV infection over the past decade. This underlines the fact that early initiation of combination antiretroviral therapy (cART), whilst minimising the size of the viral reservoir, is insufficient to achieve cure, unless other factors contribute. In this review, we consider these additional factors that may facilitate functional cure in paediatric infection. Among the early life immune activity, these include HIV-specific cytotoxic T-lymphocyte (CTL) and natural killer (NK) cell responses. The former have less potent antiviral efficacy in paediatric compared with adult infection, and indeed, in early life, NK responses have greater impact in suppressing viral replication than CTL. This fact may contribute to a greater potential for functional cure to be achieved in paediatric versus adult infection, since post-treatment control in adults is associated less with highly potent CTL activity, and more with effective antiviral NK cell responses. Nonetheless, antiviral CTL responses can play an increasingly effective role through childhood, especially in individuals expressing then 'protective' HLA-I molecules HLA-B*27/57/58:01/8101. The role of the innate system on preventing infection, in shaping the particular viruses transmitted, and influencing outcome is discussed. The susceptibility of female fetuses to in utero mother-to-child transmission, especially in the setting of recent maternal infection, is a curiosity that also provides clues to mechanisms by which cure may be achieved, since initial findings are that viral rebound is less frequent among males who interrupt cART. The potential of broadly neutralising antibody therapy to facilitate cure in children who have received early cART is discussed. Finally, we draw attention to the impact of the changing face of the paediatric HIV epidemic on cure potential. The effect of cART is not limited to preventing AIDS and reducing the risk of transmission. cART also affects which mothers transmit. No longer are mothers who transmit those who carry genes associated with poor immune control of HIV. In the cART era, a high proportion (>70% in our South African study) of transmitting mothers are those who seroconvert in pregnancy or who for social reasons are diagnosed late in pregnancy. As a result, now, genes associated with poor immune control of HIV are not enriched in mothers who transmit HIV to their child. These changes will likely influence the effectiveness of HLA-associated immune responses and therefore cure potential among children.
Collapse
Affiliation(s)
- Vinicius A. Vieira
- Peter Medawar Building for Pathogen Research, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Nicholas Herbert
- Africa Health Research Institute (AHRI), Nelson R Mandela School of Medicine, Durban, South Africa
| | - Gabriela Cromhout
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Emily Adland
- Peter Medawar Building for Pathogen Research, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Philip Goulder
- Peter Medawar Building for Pathogen Research, Department of Paediatrics, University of Oxford, Oxford, United Kingdom,Africa Health Research Institute (AHRI), Nelson R Mandela School of Medicine, Durban, South Africa,HIV Pathogenesis Programme, Doris Duke Medical Research Institute, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa,*Correspondence: Philip Goulder,
| |
Collapse
|
15
|
Mitochondria-Mediated Cardiovascular Benefits of Sodium-Glucose Co-Transporter 2 Inhibitors. Int J Mol Sci 2022; 23:ijms23105371. [PMID: 35628174 PMCID: PMC9140946 DOI: 10.3390/ijms23105371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/04/2022] [Accepted: 05/10/2022] [Indexed: 11/23/2022] Open
Abstract
Several recent cardiovascular trials of SGLT 2 (sodium-glucose cotransporter 2) inhibitors revealed that they could reduce adverse cardiovascular events in patients with T2DM (type 2 diabetes mellitus). However, the exact molecular mechanism underlying the beneficial effects that SGLT2 inhibitors have on the cardiovascular system is still unknown. In this review, we focus on the molecular mechanisms of the mitochondria-mediated beneficial effects of SGLT2 inhibitors on the cardiovascular system. The application of SGLT2 inhibitors ameliorates mitochondrial dysfunction, dynamics, bioenergetics, and ion homeostasis and reduces the production of mitochondrial reactive oxygen species, which results in cardioprotective effects. Herein, we present a comprehensive overview of the impact of SGLT2 inhibitors on mitochondria and highlight the potential application of these medications to treat both T2DM and cardiovascular diseases.
Collapse
|
16
|
Frutos-Rincón L, Gómez-Sánchez JA, Íñigo-Portugués A, Acosta MC, Gallar J. An Experimental Model of Neuro-Immune Interactions in the Eye: Corneal Sensory Nerves and Resident Dendritic Cells. Int J Mol Sci 2022; 23:ijms23062997. [PMID: 35328417 PMCID: PMC8951464 DOI: 10.3390/ijms23062997] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 02/28/2022] [Accepted: 03/04/2022] [Indexed: 12/04/2022] Open
Abstract
The cornea is an avascular connective tissue that is crucial, not only as the primary barrier of the eye but also as a proper transparent refractive structure. Corneal transparency is necessary for vision and is the result of several factors, including its highly organized structure, the physiology of its few cellular components, the lack of myelinated nerves (although it is extremely innervated), the tightly controlled hydration state, and the absence of blood and lymphatic vessels in healthy conditions, among others. The avascular, immune-privileged tissue of the cornea is an ideal model to study the interactions between its well-characterized and dense sensory nerves (easily accessible for both focal electrophysiological recording and morphological studies) and the low number of resident immune cell types, distinguished from those cells migrating from blood vessels. This paper presents an overview of the corneal structure and innervation, the resident dendritic cell (DC) subpopulations present in the cornea, their distribution in relation to corneal nerves, and their role in ocular inflammatory diseases. A mouse model in which sensory axons are constitutively labeled with tdTomato and DCs with green fluorescent protein (GFP) allows further analysis of the neuro-immune crosstalk under inflammatory and steady-state conditions of the eye.
Collapse
Affiliation(s)
- Laura Frutos-Rincón
- Instituto de Neurociencias, Universidad Miguel Hernández—Consejo Superior de Investigaciones Científicas, 03550 San Juan de Alicante, Spain; (L.F.-R.); (A.Í.-P.); (M.C.A.); (J.G.)
- The European University of Brain and Technology-NeurotechEU, 03550 San Juan de Alicante, Spain
| | - José Antonio Gómez-Sánchez
- Instituto de Neurociencias, Universidad Miguel Hernández—Consejo Superior de Investigaciones Científicas, 03550 San Juan de Alicante, Spain; (L.F.-R.); (A.Í.-P.); (M.C.A.); (J.G.)
- Correspondence: ; Tel.: +34-965-91-9594
| | - Almudena Íñigo-Portugués
- Instituto de Neurociencias, Universidad Miguel Hernández—Consejo Superior de Investigaciones Científicas, 03550 San Juan de Alicante, Spain; (L.F.-R.); (A.Í.-P.); (M.C.A.); (J.G.)
| | - M. Carmen Acosta
- Instituto de Neurociencias, Universidad Miguel Hernández—Consejo Superior de Investigaciones Científicas, 03550 San Juan de Alicante, Spain; (L.F.-R.); (A.Í.-P.); (M.C.A.); (J.G.)
- The European University of Brain and Technology-NeurotechEU, 03550 San Juan de Alicante, Spain
| | - Juana Gallar
- Instituto de Neurociencias, Universidad Miguel Hernández—Consejo Superior de Investigaciones Científicas, 03550 San Juan de Alicante, Spain; (L.F.-R.); (A.Í.-P.); (M.C.A.); (J.G.)
- The European University of Brain and Technology-NeurotechEU, 03550 San Juan de Alicante, Spain
- Instituto de Investigación Biomédica y Sanitaria de Alicante, 03010 Alicante, Spain
| |
Collapse
|
17
|
Papadopoulou G, Manoloudi E, Repousi N, Skoura L, Hurst T, Karamitros T. Molecular and Clinical Prognostic Biomarkers of COVID-19 Severity and Persistence. Pathogens 2022; 11:311. [PMID: 35335635 PMCID: PMC8948624 DOI: 10.3390/pathogens11030311] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/24/2022] [Accepted: 02/27/2022] [Indexed: 02/04/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), poses several challenges to clinicians, due to its unpredictable clinical course. The identification of laboratory biomarkers, specific cellular, and molecular mediators of immune response could contribute to the prognosis and management of COVID-19 patients. Of utmost importance is also the detection of differentially expressed genes, which can serve as transcriptomic signatures, providing information valuable to stratify patients into groups, based on the severity of the disease. The role of biomarkers such as IL-6, procalcitonin, neutrophil-lymphocyte ratio, white blood cell counts, etc. has already been highlighted in recently published studies; however, there is a notable amount of new evidence that has not been summarized yet, especially regarding transcriptomic signatures. Hence, in this review, we assess the latest cellular and molecular data and determine the significance of abnormalities in potential biomarkers for COVID-19 severity and persistence. Furthermore, we applied Gene Ontology (GO) enrichment analysis using the genes reported as differentially expressed in the literature in order to investigate which biological pathways are significantly enriched. The analysis revealed a number of processes, such as inflammatory response, and monocyte and neutrophil chemotaxis, which occur as part of the complex immune response to SARS-CoV-2.
Collapse
Affiliation(s)
- Gethsimani Papadopoulou
- Bioinformatics and Applied Genomics Unit, Department of Microbiology, Hellenic Pasteur Institute, 115 21 Athens, Greece; (G.P.); (E.M.); (N.R.)
| | - Eleni Manoloudi
- Bioinformatics and Applied Genomics Unit, Department of Microbiology, Hellenic Pasteur Institute, 115 21 Athens, Greece; (G.P.); (E.M.); (N.R.)
| | - Nikolena Repousi
- Bioinformatics and Applied Genomics Unit, Department of Microbiology, Hellenic Pasteur Institute, 115 21 Athens, Greece; (G.P.); (E.M.); (N.R.)
| | - Lemonia Skoura
- Department of Microbiology, AHEPA University Hospital, Medical School, Aristotle University of Thessaloniki, 546 36 Thessaloniki, Greece;
| | - Tara Hurst
- School of Health Sciences, Birmingham City University, Birmingham B15 3TN, UK;
| | - Timokratis Karamitros
- Bioinformatics and Applied Genomics Unit, Department of Microbiology, Hellenic Pasteur Institute, 115 21 Athens, Greece; (G.P.); (E.M.); (N.R.)
| |
Collapse
|
18
|
Bunis DG, Wang W, Vallvé-Juanico J, Houshdaran S, Sen S, Ben Soltane I, Kosti I, Vo KC, Irwin JC, Giudice LC, Sirota M. Whole-Tissue Deconvolution and scRNAseq Analysis Identify Altered Endometrial Cellular Compositions and Functionality Associated With Endometriosis. Front Immunol 2022; 12:788315. [PMID: 35069565 PMCID: PMC8766492 DOI: 10.3389/fimmu.2021.788315] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 12/09/2021] [Indexed: 12/13/2022] Open
Abstract
The uterine lining (endometrium) exhibits a pro-inflammatory phenotype in women with endometriosis, resulting in pain, infertility, and poor pregnancy outcomes. The full complement of cell types contributing to this phenotype has yet to be identified, as most studies have focused on bulk tissue or select cell populations. Herein, through integrating whole-tissue deconvolution and single-cell RNAseq, we comprehensively characterized immune and nonimmune cell types in the endometrium of women with or without disease and their dynamic changes across the menstrual cycle. We designed metrics to evaluate specificity of deconvolution signatures that resulted in single-cell identification of 13 novel signatures for immune cell subtypes in healthy endometrium. Guided by statistical metrics, we identified contributions of endometrial epithelial, endothelial, plasmacytoid dendritic cells, classical dendritic cells, monocytes, macrophages, and granulocytes to the endometrial pro-inflammatory phenotype, underscoring roles for nonimmune as well as immune cells to the dysfunctionality of this tissue.
Collapse
Affiliation(s)
- Daniel G. Bunis
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Wanxin Wang
- Center for Reproductive Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Júlia Vallvé-Juanico
- Center for Reproductive Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Sahar Houshdaran
- Center for Reproductive Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Sushmita Sen
- Center for Reproductive Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Isam Ben Soltane
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Idit Kosti
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Kim Chi Vo
- Center for Reproductive Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Juan C. Irwin
- Center for Reproductive Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Linda C. Giudice
- Center for Reproductive Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Marina Sirota
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, United States
- Department of Pediatrics, Division of Neonatology, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
19
|
Zhang W, An EK, Hwang J, Jin JO. Mice Plasmacytoid Dendritic Cells Were Activated by Lipopolysaccharides Through Toll-Like Receptor 4/Myeloid Differentiation Factor 2. Front Immunol 2021; 12:727161. [PMID: 34603298 PMCID: PMC8481683 DOI: 10.3389/fimmu.2021.727161] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 09/02/2021] [Indexed: 11/29/2022] Open
Abstract
Plasmacytoid dendritic cells (pDCs) are known to respond to viral infections. However, the activation of pDCs by bacterial components such as lipopolysaccharides (LPS) has not been well studied. Here, we found that pDCs, conventional dendritic cells (cDCs), and B cells express high levels of toll-like receptor 4 (TLR4), a receptor for LPS. Moreover, LPS could effectively bind to not only cDCs but also pDCs and B cells. Intraperitoneal administration of LPS promoted activation of splenic pDCs and cDCs. LPS treatment led to upregulation of interferon regulatory factor 7 (IRF7) and induced production of interferon-alpha (IFN-α) in splenic pDCs. Furthermore, LPS-dependent upregulation of co-stimulatory molecules in pDCs did not require the assistance of other immune cells, such as cDCs. However, the production levels of IFN-α were decreased in cDC-depleted splenocytes, indicating that cDCs may contribute to the enhancement of IFN-α production in pDCs. Finally, we showed that activation of pDCs by LPS requires the TLR4 and myeloid differentiation factor 2 (MD2) signaling pathways. Thus, these results demonstrate that the gram-negative component LPS can directly stimulate pDCs via TLR4/MD2 stimulation in mice.
Collapse
Affiliation(s)
- Wei Zhang
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Eun-Koung An
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, South Korea
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Juyoung Hwang
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, China
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, South Korea
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Jun-O Jin
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, China
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, South Korea
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| |
Collapse
|
20
|
Seo SU, Jeong JH, Baek BS, Choi JM, Choi YS, Ko HJ, Kweon MN. Bleomycin-Induced Lung Injury Increases Resistance to Influenza Virus Infection in a Type I Interferon-Dependent Manner. Front Immunol 2021; 12:697162. [PMID: 34484196 PMCID: PMC8416411 DOI: 10.3389/fimmu.2021.697162] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 07/29/2021] [Indexed: 01/07/2023] Open
Abstract
Acute lung injury (ALI) results in acute respiratory disease that causes fatal respiratory diseases; however, little is known about the incidence of influenza infection in ALI. Using a ALI-mouse model, we investigated the pro-inflammatory cytokine response to ALI and influenza infection. Mice treated with bleomycin (BLM), which induces ALI, were more resistant to influenza virus infection and exhibited higher levels of type I interferon (IFN-I) transcription during the early infection period than that in PBS-treated control mice. BLM-treated mice also exhibited a lower viral burden, reduced pro-inflammatory cytokine production, and neutrophil levels. In contrast, BLM-treated IFN-I receptor 1 (IFNAR1)-knockout mice failed to show this attenuated phenotype, indicating that IFN-I is key to the antiviral response in ALI-induced mice. The STING/TBK1/IRF3 pathway was found to be involved in IFN-I production and the establishment of an antiviral environment in the lung. The depletion of plasmacytoid dendritic cells (pDCs) reduced the effect of BLM treatment against influenza virus infection, suggesting that pDCs are the major source of IFN-I and are crucial for defense against viral infection in BLM-induced lung injury. Overall, this study showed that BLM-mediated ALI in mice induced the release of double-stranded DNA, which in turn potentiated IFN-I-dependent pulmonary viral resistance by activating the STING/TBK1/IRF3 pathway in association with pDCs.
Collapse
Affiliation(s)
- Sang-Uk Seo
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jae-Hyeon Jeong
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, Chuncheon, South Korea
| | - Bum-Seo Baek
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.,Wide River Institute of Immunology, Seoul National University College of Medicine, Hongcheon, South Korea
| | - Je-Min Choi
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, South Korea
| | - Youn Soo Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| | - Hyun-Jeong Ko
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, Chuncheon, South Korea
| | - Mi-Na Kweon
- Mucosal Immunology Laboratory, Department of Convergence Medicine, University of Ulsan College of Medicine/Asan Medical Center, Seoul, South Korea
| |
Collapse
|
21
|
Illouz T, Biragyn A, Iulita MF, Flores-Aguilar L, Dierssen M, De Toma I, Antonarakis SE, Yu E, Herault Y, Potier MC, Botté A, Roper R, Sredni B, London J, Mobley W, Strydom A, Okun E. Immune Dysregulation and the Increased Risk of Complications and Mortality Following Respiratory Tract Infections in Adults With Down Syndrome. Front Immunol 2021; 12:621440. [PMID: 34248930 PMCID: PMC8267813 DOI: 10.3389/fimmu.2021.621440] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 06/02/2021] [Indexed: 12/12/2022] Open
Abstract
The risk of severe outcomes following respiratory tract infections is significantly increased in individuals over 60 years, especially in those with chronic medical conditions, i.e., hypertension, diabetes, cardiovascular disease, dementia, chronic respiratory disease, and cancer. Down Syndrome (DS), the most prevalent intellectual disability, is caused by trisomy-21 in ~1:750 live births worldwide. Over the past few decades, a substantial body of evidence has accumulated, pointing at the occurrence of alterations, impairments, and subsequently dysfunction of the various components of the immune system in individuals with DS. This associates with increased vulnerability to respiratory tract infections in this population, such as the influenza virus, respiratory syncytial virus, SARS-CoV-2 (COVID-19), and bacterial pneumonias. To emphasize this link, here we comprehensively review the immunobiology of DS and its contribution to higher susceptibility to severe illness and mortality from respiratory tract infections.
Collapse
Affiliation(s)
- Tomer Illouz
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan, Israel
- The Paul Feder Laboratory on Alzheimer’s Disease Research, Bar-Ilan University, Ramat Gan, Israel
| | - Arya Biragyn
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institute of Health, Baltimore, MD, United States
| | - Maria Florencia Iulita
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Lisi Flores-Aguilar
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| | - Mara Dierssen
- Center for Genomic Regulation, The Barcelona Institute for Science and Technology, Barcelona, Spain
- University Pompeu Fabra, Barcelona, Spain
- Biomedical Research Networking Center for Rare Diseases (CIBERER), Barcelona, Spain
| | - Ilario De Toma
- Center for Genomic Regulation, The Barcelona Institute for Science and Technology, Barcelona, Spain
- University Pompeu Fabra, Barcelona, Spain
- Biomedical Research Networking Center for Rare Diseases (CIBERER), Barcelona, Spain
| | - Stylianos E. Antonarakis
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland
- Medigenome, Swiss Institute of Genomic Medicine, Geneva, Switzerland
- iGE3 Institute of Genetics and Genomics of Geneva, Geneva, Switzerland
| | - Eugene Yu
- The Children’s Guild Foundation Down Syndrome Research Program, Genetics and Genomics Program and Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
- Genetics, Genomics and Bioinformatics Program, State University of New York at Buffalo, Buffalo, NY, United States
| | - Yann Herault
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique Biologie Moléculaire et Cellulaire, IGBMC - UMR 7104 - Inserm U1258, Illkirch, France
| | - Marie-Claude Potier
- Paris Brain Institute (ICM), CNRS UMR7225, INSERM U1127, Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Alexandra Botté
- Paris Brain Institute (ICM), CNRS UMR7225, INSERM U1127, Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Randall Roper
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| | - Benjamin Sredni
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | | | - William Mobley
- Department of Neurosciences, University of California, San Diego, San Diego, CA, United States
| | - Andre Strydom
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry Psychology and Neuroscience, King’s College London, London, United Kingdom
- South London and Maudsley NHS Foundation Trust, London, United Kingdom
| | - Eitan Okun
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan, Israel
- The Paul Feder Laboratory on Alzheimer’s Disease Research, Bar-Ilan University, Ramat Gan, Israel
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| |
Collapse
|
22
|
Zengin HB, Pukhalskaya T, Smoller BR. Role of CD123 (+) Plasmacytoid Dendritic Cells in Etiologically Different Variants of Erythema Multiforme: A Monocentric Retrospective Study. Dermatopathology (Basel) 2021; 8:89-96. [PMID: 33916862 PMCID: PMC8167774 DOI: 10.3390/dermatopathology8020014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 03/31/2021] [Accepted: 04/01/2021] [Indexed: 11/17/2022] Open
Abstract
Plasmacytoid dendritic cells (pDCs) constitute a subset of dendritic cells known to be the “professional” interferon type I (IFN-I) producers. pDCs play an important role in antiviral immunity, as well as linking innate and adaptive immunity. Under normal conditions pDCs are not present in skin. They are shown to be a part of the inflammatory infiltrate in different skin conditions including erythema multiforme (EM). This condition is considered to be a cell-mediated immune reaction to a wide variety of agents, most commonly herpes simplex virus. Nevertheless, the pathophysiology of EM still remains unclear. In this study, we grouped 32 biopsies from 30 patients diagnosed with EM, based on their etiology and analyzed the density and distribution of CD123 positive pDCs. In all cases we observed a greatly increased number of pDCs in the dermal inflammatory infiltrate. Virally-induced EM (by herpes simplex virus (HSV) and other viruses) was more likely to have a significantly higher number of pDCs compared to non-virally associated EM. Hence, we think that pDCs play a key role in the pathogenesis of EM independent of etiology and may play an increased role in virally-associated cases. Further studies on pDCs would clarify their importance in EM and improve our understanding of the pathophysiology of this disease.
Collapse
|
23
|
Pathak M, Lal G. The Regulatory Function of CCR9 + Dendritic Cells in Inflammation and Autoimmunity. Front Immunol 2020; 11:536326. [PMID: 33123124 PMCID: PMC7566413 DOI: 10.3389/fimmu.2020.536326] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 08/13/2020] [Indexed: 12/26/2022] Open
Abstract
Chemokine receptor CCR9 is a G protein–coupled receptor and expressed on several types of immune cells, including dendritic cells (DCs), CD4+ T cells, and B cells. CCR9 drives the migration of immune cells to gradients of its cognate ligand CCL25. The chemokine CCL25 is mostly produced by gut and thymic epithelial cells. Gut- and thymic-homing DCs are known to express CCR9, and these cells are predominantly localized in the gut lining and thymus. CCR9+ DCs are implicated in regulating inflammation, food allergy, alloimmunity, and autoimmunity. Differential interaction of CCR9+ DCs with lymphoid and myeloid cells in the thymus, secondary lymphoid tissues, and mucosal sites offer crucial insights to immune regulation. In this review, we examine the phenotypes, distributions, and interactions of CCR9+ DCs with other immune cells, elucidating their functions and role in inflammation and autoimmunity.
Collapse
Affiliation(s)
- Manisha Pathak
- Laboratory of Autoimmunity and Tolerance, National Centre for Cell Science, Pune, India
| | - Girdhari Lal
- Laboratory of Autoimmunity and Tolerance, National Centre for Cell Science, Pune, India
| |
Collapse
|
24
|
Eixarch H, Calvo-Barreiro L, Costa C, Reverter-Vives G, Castillo M, Gil V, Del Río JA, Montalban X, Espejo C. Inhibition of the BMP Signaling Pathway Ameliorated Established Clinical Symptoms of Experimental Autoimmune Encephalomyelitis. Neurotherapeutics 2020; 17:1988-2003. [PMID: 32681355 PMCID: PMC7851289 DOI: 10.1007/s13311-020-00885-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) are secreted growth factors that belong to the transforming growth factor beta superfamily. BMPs have been implicated in physiological processes, but they are also involved in many pathological conditions. Multiple sclerosis (MS) is an immune-mediated disease of the central nervous system (CNS); however, its etiology remains elusive. Some evidence points to BMPs as important players in the pathogenesis of inflammatory and autoimmune disorders. In the present work, we studied the expression of BMP2, BMP4, BMP5, BMP6, BMP7, BMP type II receptor, and noggin in the immune system during different phases of experimental autoimmune encephalomyelitis (EAE). Major changes in the expression of BMPs took place in the initial phases of EAE. Indeed, those changes mainly affected BMP6 (whose expression was abrogated), BMP2, and BMP7 (whose expression was increased). In addition, we showed that in vivo inhibition of the BMP signaling pathway with small molecules ameliorated the already established clinical symptoms of EAE, as well as the CNS histopathological features. At the immune level, we observed an expansion of plasmacytoid dendritic cells (pDCs) in mice treated with small molecules that inhibit the BMP signaling pathway. pDCs could play an important role in promoting the expansion of antigen-specific regulatory T cells. Altogether, our data suggest a role for BMPs in early immune events that take place in myelin oligodendrocyte glycoprotein (MOG)-induced EAE. In addition, the clinical outcome of the disease was improved when the BMP signaling pathway was inhibited in mice that presented established EAE symptoms.
Collapse
Affiliation(s)
- Herena Eixarch
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya, Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Universitat Autònoma de Barcelona, 08193, Bellaterra (Cerdanyola del Vallès), Spain
| | - Laura Calvo-Barreiro
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya, Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Universitat Autònoma de Barcelona, 08193, Bellaterra (Cerdanyola del Vallès), Spain
| | - Carme Costa
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya, Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Universitat Autònoma de Barcelona, 08193, Bellaterra (Cerdanyola del Vallès), Spain
| | - Gemma Reverter-Vives
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya, Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Universitat Autònoma de Barcelona, 08193, Bellaterra (Cerdanyola del Vallès), Spain
| | - Mireia Castillo
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya, Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Universitat Autònoma de Barcelona, 08193, Bellaterra (Cerdanyola del Vallès), Spain
| | - Vanessa Gil
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Parc Científic de Barcelona, Barcelona, Spain
- Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - José Antonio Del Río
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Parc Científic de Barcelona, Barcelona, Spain
- Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Xavier Montalban
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya, Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Universitat Autònoma de Barcelona, 08193, Bellaterra (Cerdanyola del Vallès), Spain
- Division of Neurology, St Michael's Hospital, University of Toronto, Toronto, Canada
| | - Carmen Espejo
- Servei de Neurologia-Neuroimmunologia, Centre d'Esclerosi Múltiple de Catalunya, Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Barcelona, Spain.
- Universitat Autònoma de Barcelona, 08193, Bellaterra (Cerdanyola del Vallès), Spain.
| |
Collapse
|
25
|
Alumina Ceramic Exacerbates the Inflammatory Disease by Activation of Macrophages and T Cells. Int J Mol Sci 2020; 21:ijms21197114. [PMID: 32993182 PMCID: PMC7583733 DOI: 10.3390/ijms21197114] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/15/2020] [Accepted: 09/24/2020] [Indexed: 11/16/2022] Open
Abstract
(1) Background: Aluminum oxide (Al2O3) ceramic is one of the materials used for artificial joints, and it has been known that their fine particles (FPs) are provided by the wear of the ceramic. Al2O3 FPs have been shown to induce macrophage activation in vitro; however, the inflammatory effect in vivo has not been studied. (2) Methods: We examined the in vivo effect of Al2O3 FPs on the innate and adaptive immune cells in the mice. (3) Results: Al2O3 FPs promoted the activation of spleen macrophages; however, conventional dendritic cells (cDCs), plasmacytoid DCs (pDCs), and natural killer (NK) cells were not activated. In addition, increases in the CD4 and CD8 T cells was induced in the spleens of the mice treated with Al2O3 FPs, which differentiated into interferon-gamma (IFN-γ)-producing helper T1 (Th1) and cytotoxic T1 (Tc1) cells. Finally, the injection of Al2O3 FPs exacerbated dextran sulfate sodium (DSS)-induced inflammation in the colon, mediated by activated and increased number of CD4 and CD8 T cells. (4) Conclusions: These data demonstrate that FPs of Al2O3 ceramic may contribute to the exacerbation of inflammatory diseases in the patients.
Collapse
|
26
|
Zhang W, Lim SM, Hwang J, Ramalingam S, Kim M, Jin JO. Monophosphoryl lipid A-induced activation of plasmacytoid dendritic cells enhances the anti-cancer effects of anti-PD-L1 antibodies. Cancer Immunol Immunother 2020; 70:689-700. [PMID: 32902663 DOI: 10.1007/s00262-020-02715-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 08/31/2020] [Indexed: 12/23/2022]
Abstract
Monophosphoryl lipid A (MPLA) is a toll-like receptor 4 ligand that promotes immune activation in mice and humans, without undesired inflammation. Immunotherapy by the combining immune checkpoint blockade and MPLA has shown promising anti-cancer effects in both mice and humans. In this study, we explored how MPLA enhanced the anti-cancer effects of anti-PD-L1 antibodies (Abs). Anti-cancer immunity induced by the combination of anti-PD-L1 Abs and MPLA failed in CD4 and CD8 cell-depleted mice. Moreover, the combination treatment of anti-PD-L1 Abs and MPLA synergistically enhanced the activation of plasmacytoid dendritic cells (pDCs) in the mouse in vivo, while conventional DCs were not. In addition, mice treated with anti-PD-L1 Abs and MPLA were not protected from B16 melanoma by blockade of interferon-alpha receptor (IFNAR). The combination of anti-PD-L1 Abs and MPLA also promoted human peripheral blood pDC activation and induced IFN-α-dependent T cell activation. Therefore, these results demonstrate that MPLA enhances anti-PD-L1 Ab-mediated anti-cancer immunity through the activation and IFN-α production of pDCs.
Collapse
Affiliation(s)
- Wei Zhang
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, 201508, China
| | - Seong-Min Lim
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, Republic of Korea.,Research Institute of Cell Culture, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Juyoung Hwang
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, 201508, China.,Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, Republic of Korea.,Research Institute of Cell Culture, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Srinivasan Ramalingam
- Department of Food Science and Technology, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Myunghee Kim
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, 38541, Republic of Korea.,Department of Food Science and Technology, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Jun-O Jin
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, 201508, China. .,Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, Republic of Korea. .,Research Institute of Cell Culture, Yeungnam University, Gyeongsan, 38541, Republic of Korea.
| |
Collapse
|
27
|
Chen HJ, Tas SW, de Winther MPJ. Type-I interferons in atherosclerosis. J Exp Med 2020; 217:132613. [PMID: 31821440 PMCID: PMC7037237 DOI: 10.1084/jem.20190459] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 10/05/2019] [Accepted: 10/30/2019] [Indexed: 12/13/2022] Open
Abstract
Chen et al. review the effects of type-I IFNs and the potential of anti–type-I IFN therapies in atherosclerosis. The contribution of dyslipidemia and inflammation in atherosclerosis is well established. Along with effective lipid-lowering treatments, the recent success of clinical trials with anti-inflammatory therapies and the accelerated atherosclerosis in many autoimmune diseases suggest that targeting inflammation may open new avenues for the prevention and the treatment for cardiovascular diseases (CVDs). In the past decades, studies have widened the role of type-I interferons (IFNs) in disease, from antivirus defense to autoimmune responses and immuno-metabolic syndromes. While elevated type-I IFN level in serum is associated with CVD incidence in patients with interferonopathies, experimental data have attested that type-I IFNs affect plaque-residing macrophages, potentiate foam cell and extracellular trap formation, induce endothelial dysfunction, alter the phenotypes of dendritic cells and T and B lymphocytes, and lead to exacerbated atherosclerosis outcomes. In this review, we discuss the production and the effects of type-I IFNs in different atherosclerosis-associated cell types from molecular biology studies, animal models, and clinical observations, and the potential of new therapies against type-I IFN signaling for atherosclerosis.
Collapse
Affiliation(s)
- Hung-Jen Chen
- Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Sander W Tas
- Amsterdam Rheumatology and Immunology Center, Department of Rheumatology and Clinical Immunology, and Laboratory for Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, Netherlands
| | - Menno P J de Winther
- Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands.,Institute for Cardiovascular Prevention, Ludwig Maximilians University, Munich, Germany
| |
Collapse
|
28
|
Combination of Mycobacterium indicus pranii and Heat-Induced Promastigotes Cures Drug-Resistant Leishmania Infection: Critical Role of Interleukin-6-Producing Classical Dendritic Cells. Infect Immun 2020; 88:IAI.00222-19. [PMID: 32229617 DOI: 10.1128/iai.00222-19] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 03/24/2020] [Indexed: 12/18/2022] Open
Abstract
The major issues in available therapeutic modalities against leishmaniasis are cost, toxicity, and the emergence of drug resistance. The aim of this work was to develop a successful therapeutic adjuvant against drug-resistant Leishmania donovani infection by means of combining Mycobacterium indicus pranii with heat-induced promastigotes (HIP). One-month postinfected BALB/c mice were administered subcutaneously with M. indicus pranii (108 cells) and HIP (100 μg) for 5 days. Spleens were harvested for flow cytometric and reverse transcriptase PCR analysis. The antileishmanial effect of the combination strategy was associated with induction of a disease-resolving Th1 and Th17 response with simultaneous downregulation of CD4+ CD25+ Foxp3+ (nTreg) cells and CD4+ CD25- Foxp3- (Tr1) cells in the spleen. The significant expansion of CD4+ TCM (CD4+ CD44hi CD11ahi CD62Lhi) cells was a further interesting outcome of this therapeutic strategy in the context of long-term protection of hosts against secondary infection. Toll-like receptor 2 (TLR2) was also found instrumental in this antiparasitic therapy. Induced interleukin-6 (IL-6) production from expanded CD11c+ CD8α+ (cDC1) and CD11c+ CD11b+ (cDC2) dendritic cells (DCs) but not from the CD11b+ Ly6c+ inflammatory monocytes (iMOs), was found critical in the protective expansion of Th17 as evidenced by an in vivo IL-6 neutralization assay. It also promoted the hematopoietic conversion toward DC progenitors (pre-DCs) from common dendritic cell progenitors (CDPs), the immediate precursors, in bone marrow. This novel combinational strategy demonstrated that expansion of Th17 by IL-6 released from CD11c+ classical DCs is crucial, together with the conventional Th1 response, to control drug-resistant infection.
Collapse
|
29
|
Li B, Li Y, Li K, Zhu L, Yu Q, Cai P, Fang J, Zhang W, Du P, Jiang C, Lin J, Qu K. APEC: an accesson-based method for single-cell chromatin accessibility analysis. Genome Biol 2020; 21:116. [PMID: 32398051 PMCID: PMC7218568 DOI: 10.1186/s13059-020-02034-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 04/30/2020] [Indexed: 12/21/2022] Open
Abstract
The development of sequencing technologies has promoted the survey of genome-wide chromatin accessibility at single-cell resolution. However, comprehensive analysis of single-cell epigenomic profiles remains a challenge. Here, we introduce an accessibility pattern-based epigenomic clustering (APEC) method, which classifies each cell by groups of accessible regions with synergistic signal patterns termed “accessons”. This python-based package greatly improves the accuracy of unsupervised single-cell clustering for many public datasets. It also predicts gene expression, identifies enriched motifs, discovers super-enhancers, and projects pseudotime trajectories. APEC is available at https://github.com/QuKunLab/APEC.
Collapse
Affiliation(s)
- Bin Li
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Young Li
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Kun Li
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Lianbang Zhu
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Qiaoni Yu
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Pengfei Cai
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Jingwen Fang
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, 230001, Anhui, China.,HanGene Biotech, Xiaoshan Innovation Polis, Hangzhou, 310000, Zhejiang, China
| | - Wen Zhang
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Pengcheng Du
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Chen Jiang
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Jun Lin
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Kun Qu
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, 230001, Anhui, China. .,CAS Center for Excellence in Molecular Cell Sciences, The CAS Key Laboratory of Innate Immunity and Chronic Disease, University of Science and Technology of China, Hefei, 230027, Anhui, China.
| |
Collapse
|
30
|
Abstract
Sexual dimorphisms account for differences in clinical manifestations or incidence of infectious or autoimmune diseases and malignancy between females and males. Females develop enhanced innate and adaptive immune responses than males and are less susceptible to many infections of bacterial, viral, parasitic, and fungal origin and malignancies but in contrast, they are more prone to develop autoimmune diseases. The higher susceptibility to infections in males is observed from birth to adulthood, suggesting that sex chromosomes and not sex hormones have a major role in sexual dimorphism in innate immunity. Sex-based regulation of immune responses ultimately contributes to age-related disease development and life expectancy. Differences between males and females have been described in the expression of pattern recognition receptors of the innate immune response and in the functional responses of phagocytes and antigen presenting cells. Different factors have been shown to account for the sex-based disparity in immune responses, including genetic factors and hormonal mediators, which contribute independently to dimorphism in the innate immune response. For instance, several genes encoding for innate immune molecules are located on the X chromosome. In addition, estrogen and/or testosterone have been reported to modulate the differentiation, maturation, lifespan, and effector functions of innate immune cells, including neutrophils, macrophages, natural killer cells, and dendritic cells. In this review, we will focus on differences between males and females in innate immunity, which represents the first line of defense against pathogens and plays a fundamental role in the activation, regulation, and orientation of the adaptive immune response.
Collapse
Affiliation(s)
- Sébastien Jaillon
- Humanitas University, Department of Biomedical Sciences, Via Rita Levi Montalcini, 20090, Pieve Emanuele, Milan, Italy. .,Humanitas Clinical and Research Center, Via Manzoni 56, 20089, Rozzano, Milan, Italy.
| | - Kevin Berthenet
- Humanitas Clinical and Research Center, Via Manzoni 56, 20089, Rozzano, Milan, Italy
| | - Cecilia Garlanda
- Humanitas University, Department of Biomedical Sciences, Via Rita Levi Montalcini, 20090, Pieve Emanuele, Milan, Italy. .,Humanitas Clinical and Research Center, Via Manzoni 56, 20089, Rozzano, Milan, Italy.
| |
Collapse
|
31
|
Chistiakov DA, Kashirskikh DA, Khotina VA, Grechko AV, Orekhov AN. Immune-Inflammatory Responses in Atherosclerosis: The Role of Myeloid Cells. J Clin Med 2019; 8:jcm8111798. [PMID: 31717832 PMCID: PMC6912749 DOI: 10.3390/jcm8111798] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 10/24/2019] [Indexed: 01/28/2023] Open
Abstract
Inflammation plays a key role in the initiation and progression of atherosclerosis and can be caused by multiple agents, including increased concentration of circulating low-density lipoprotein (LDL) cholesterol. Areas of the arterial wall affected by atherosclerosis are enriched with lymphocytes and dendritic cells (DCs). Atherosclerotic plaques contain a variety of proinflammatory immune cells, such as macrophages, DCs, T cells, natural killer cells, neutrophils and others. Intracellular lipid accumulation in atherosclerotic plaque leads to formation of so-called foam cells, the cytoplasm of which is filled with lipid droplets. According to current understanding, these cells can also derive from the immune cells that engulf lipids by means of phagocytosis. Macrophages play a crucial role in the initial stages of atherogenesis by engulfing oxidized LDL (oxLDL) in the intima that leads to their transformation to foam cells. Dying macrophages inside the plaque form a necrotic core that further aggravates the lesion. Proinflammatory DCs prime differentiation of naïve T cells to proinflammatory Th1 and Th17 subsets. In this review, we discuss the roles of cell types of myeloid origin in atherosclerosis-associated inflammation.
Collapse
Affiliation(s)
- Dimitry A. Chistiakov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia; (D.A.C.); (D.A.K.); (V.A.K.)
| | - Dmitry A. Kashirskikh
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia; (D.A.C.); (D.A.K.); (V.A.K.)
| | - Victoriya A. Khotina
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia; (D.A.C.); (D.A.K.); (V.A.K.)
| | - Andrey V. Grechko
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 109240 Moscow, Russia;
| | - Alexander N. Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia; (D.A.C.); (D.A.K.); (V.A.K.)
- Institute of Human Morphology, Tsyrupa st. 3, 117418 Moscow, Russia
- Correspondence: ; Tel.: +7-903-169-08-66
| |
Collapse
|
32
|
De Laere M, Berneman ZN, Cools N. To the Brain and Back: Migratory Paths of Dendritic Cells in Multiple Sclerosis. J Neuropathol Exp Neurol 2019; 77:178-192. [PMID: 29342287 PMCID: PMC5901086 DOI: 10.1093/jnen/nlx114] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Migration of dendritic cells (DC) to the central nervous system (CNS) is a critical event in the pathogenesis of multiple sclerosis (MS). While up until now, research has mainly focused on the transmigration of DC through the blood-brain barrier, experimental evidence points out that also the choroid plexus and meningeal vessels represent important gateways to the CNS, especially in early disease stages. On the other hand, DC can exit the CNS to maintain immunological tolerance to patterns expressed in the CNS, a process that is perturbed in MS. Targeting trafficking of immune cells, including DC, to the CNS has demonstrated to be a successful strategy to treat MS. However, this approach is known to compromise protective immune surveillance of the brain. Unravelling the migratory paths of regulatory and pathogenic DC within the CNS may ultimately lead to the design of new therapeutic strategies able to selectively interfere with the recruitment of pathogenic DC to the CNS, while leaving host protective mechanisms intact.
Collapse
Affiliation(s)
- Maxime De Laere
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp
| | - Zwi N Berneman
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp.,Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital (UZA), Edegem, Belgium
| | - Nathalie Cools
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp
| |
Collapse
|
33
|
Abstract
Immunoglobulin E (IgE), though constitutively present at low levels, is most commonly studied in atopic disease where it plays a vital role in mast cell degranulation and in initiating a T helper 2 (Th2) response. With the advent of better detection assays, however, researchers are discovering the importance of IgE in actively contributing to many disease states and pathologies. This review will discuss the latest findings in IgE beyond its role in allergies and recently discovered roles for IgE in its cell-bound form on FcεRI-expressing effector cells like monocytes and dendritic cells. In terms of parasites, we will discuss helminth-induced IgE that appears to protect the worms from immune recognition and a tick-borne illness that elicits an IgE response against red meat. Next, we describe recent findings of how auto-reactive IgE can contribute to the progression of lupus and induce organ damage. Finally, we summarize the emerging roles of IgE in tumor surveillance and antibody-dependent cytotoxicity. We additionally discuss recent or ongoing clinical trials that either target harmful IgE or use the unique characteristics of the isotype.
Collapse
Affiliation(s)
- Andrea J Luker
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA, USA
| | - Joseph C Lownik
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA, USA.,Center for Clinical and Translational Research, Virginia Commonwealth University, Richmond, VA, USA
| | - Daniel H Conrad
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA, USA
| | - Rebecca K Martin
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
34
|
Słomka A, Urban SK, Lukacs-Kornek V, Żekanowska E, Kornek M. Large Extracellular Vesicles: Have We Found the Holy Grail of Inflammation? Front Immunol 2018; 9:2723. [PMID: 30619239 PMCID: PMC6300519 DOI: 10.3389/fimmu.2018.02723] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Accepted: 11/05/2018] [Indexed: 12/17/2022] Open
Abstract
The terms microparticles (MPs) and microvesicles (MVs) refer to large extracellular vesicles (EVs) generated from a broad spectrum of cells upon its activation or death by apoptosis. The unique surface antigens of MPs/MVs allow for the identification of their cellular origin as well as its functional characterization. Two basic aspects of MP/MV functions in physiology and pathological conditions are widely considered. Firstly, it has become evident that large EVs have strong procoagulant properties. Secondly, experimental and clinical studies have shown that MPs/MVs play a crucial role in the pathophysiology of inflammation-associated disorders. A cardinal feature of these disorders is an enhanced generation of platelets-, endothelial-, and leukocyte-derived EVs. Nevertheless, anti-inflammatory effects of miscellaneous EV types have also been described, which provided important new insights into the large EV-inflammation axis. Advances in understanding the biology of MPs/MVs have led to the preparation of this review article aimed at discussing the association between large EVs and inflammation, depending on their cellular origin.
Collapse
Affiliation(s)
- Artur Słomka
- Department of Pathophysiology, Nicolaus Copernicus University in Toruń, Ludwik Rydygier Collegium Medicum, Bydgoszcz, Poland
| | - Sabine Katharina Urban
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Veronika Lukacs-Kornek
- Institute of Experimental Immunology, University Hospital of the Rheinische Friedrich-Wilhelms-University, Bonn, Germany
| | - Ewa Żekanowska
- Department of Pathophysiology, Nicolaus Copernicus University in Toruń, Ludwik Rydygier Collegium Medicum, Bydgoszcz, Poland
| | - Miroslaw Kornek
- Department of Oncology, Hematology and Rheumatology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
35
|
Pustylnikov S, Costabile F, Beghi S, Facciabene A. Targeting mitochondria in cancer: current concepts and immunotherapy approaches. Transl Res 2018; 202:35-51. [PMID: 30144423 PMCID: PMC6456045 DOI: 10.1016/j.trsl.2018.07.013] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 07/24/2018] [Accepted: 07/25/2018] [Indexed: 12/12/2022]
Abstract
An essential advantage during eukaryotic cell evolution was the acquisition of a network of mitochondria as a source of energy for cell metabolism and contrary to conventional wisdom, functional mitochondria are essential for the cancer cell. Multiple aspects of mitochondrial biology beyond bioenergetics support transformation including mitochondrial biogenesis, fission and fusion dynamics, cell death susceptibility, oxidative stress regulation, metabolism, and signaling. In cancer, the metabolism of cells is reprogrammed for energy generation from oxidative phosphorylation to aerobic glycolysis and impacts cancer mitochondrial function. Furthermore cancer cells can also modulate energy metabolism within the cancer microenvironment including immune cells and induce "metabolic anergy" of antitumor immune response. Classical approaches targeting the mitochondria of cancer cells usually aim at inducing changing energy metabolism or directly affecting functions of mitochondrial antiapoptotic proteins but most of such approaches miss the required specificity of action and carry important side effects. Several types of cancers harbor somatic mitochondrial DNA mutations and specific immune response to mutated mitochondrial proteins has been observed. An attractive alternative way to target the mitochondria in cancer cells is the induction of an adaptive immune response against mutated mitochondrial proteins. Here, we review the cancer cell-intrinsic and cell-extrinsic mechanisms through which mitochondria influence all steps of oncogenesis, with a focus on the therapeutic potential of targeting mitochondrial DNA mutations or Tumor Associated Mitochondria Antigens using the immune system.
Collapse
Affiliation(s)
- Sergey Pustylnikov
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Francesca Costabile
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Silvia Beghi
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Andrea Facciabene
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
36
|
Halpern KB, Shenhav R, Massalha H, Toth B, Egozi A, Massasa EE, Medgalia C, David E, Giladi A, Moor AE, Porat Z, Amit I, Itzkovitz S. Paired-cell sequencing enables spatial gene expression mapping of liver endothelial cells. Nat Biotechnol 2018; 36:962-970. [PMID: 30222169 PMCID: PMC6546596 DOI: 10.1038/nbt.4231] [Citation(s) in RCA: 222] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 07/19/2018] [Indexed: 02/08/2023]
Abstract
Spatially resolved single-cell RNA sequencing (scRNAseq) is a powerful approach for inferring connections between a cell's identity and its position in a tissue. We recently combined scRNAseq with spatially mapped landmark genes to infer the expression zonation of hepatocytes. However, determining zonation of small cells with low mRNA content, or without highly expressed landmark genes, remains challenging. Here we used paired-cell sequencing, in which mRNA from pairs of attached mouse cells were sequenced and gene expression from one cell type was used to infer the pairs' tissue coordinates. We applied this method to pairs of hepatocytes and liver endothelial cells (LECs). Using the spatial information from hepatocytes, we reconstructed LEC zonation and extracted a landmark gene panel that we used to spatially map LEC scRNAseq data. Our approach revealed the expression of both Wnt ligands and the Dkk3 Wnt antagonist in distinct pericentral LEC sub-populations. This approach can be used to reconstruct spatial expression maps of non-parenchymal cells in other tissues.
Collapse
Affiliation(s)
- Keren Bahar Halpern
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Rom Shenhav
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Hassan Massalha
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Beata Toth
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Adi Egozi
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Efi E. Massasa
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Chiara Medgalia
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Eyal David
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Amir Giladi
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Andreas E. Moor
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ziv Porat
- The Flow Cytometry Unit, Life Sciences Faculty, Weizmann Institute of Science, Rehovot, Israel
| | - Ido Amit
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Shalev Itzkovitz
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
37
|
Li S, Wu J, Zhu S, Liu YJ, Chen J. Disease-Associated Plasmacytoid Dendritic Cells. Front Immunol 2017; 8:1268. [PMID: 29085361 PMCID: PMC5649186 DOI: 10.3389/fimmu.2017.01268] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 09/22/2017] [Indexed: 12/20/2022] Open
Abstract
Plasmacytoid dendritic cells (pDCs), also called natural interferon (IFN)-producing cells, represent a specialized cell type within the innate immune system. pDCs are specialized in sensing viral RNA and DNA by toll-like receptor-7 and -9 and have the ability to rapidly produce massive amounts of type 1 IFNs upon viral encounter. After producing type 1 IFNs, pDCs differentiate into professional antigen-presenting cells, which are capable of stimulating T cells of the adaptive immune system. Chronic activation of human pDCs by self-DNA or mitochondrial DNA contributes to the pathogenesis of systemic lupus erythematosis and IFN-related autoimmune diseases. Under steady-state conditions, pDCs play an important role in immune tolerance. In many types of human cancers, recruitment of pDCs to the tumor microenvironment contributes to the induction of immune tolerance. Here, we provide a systemic review of recent progress in studies on the role of pDCs in human diseases, including cancers and autoimmune/inflammatory diseases.
Collapse
Affiliation(s)
- Shuang Li
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China
| | - Jing Wu
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China
| | - Shan Zhu
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China
| | - Yong-Jun Liu
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China.,Sanofi Research and Development, Cambridge, MA, United States
| | - Jingtao Chen
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China
| |
Collapse
|
38
|
Saas P, Varin A, Perruche S, Ceroi A. Recent insights into the implications of metabolism in plasmacytoid dendritic cell innate functions: Potential ways to control these functions. F1000Res 2017; 6:456. [PMID: 28580131 PMCID: PMC5437952 DOI: 10.12688/f1000research.11332.2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/19/2017] [Indexed: 12/17/2022] Open
Abstract
There are more and more data concerning the role of cellular metabolism in innate immune cells, such as macrophages or conventional dendritic cells. However, few data are available currently concerning plasmacytoid dendritic cells (PDC), another type of innate immune cells. These cells are the main type I interferon (IFN) producing cells, but they also secrete other pro-inflammatory cytokines (e.g., tumor necrosis factor or interleukin [IL]-6) or immunomodulatory factors (e.g., IL-10 or transforming growth factor-β). Through these functions, PDC participate in antimicrobial responses or maintenance of immune tolerance, and have been implicated in the pathophysiology of several autoimmune diseases, as well as in tumor immune escape mechanisms. Recent data support the idea that the glycolytic pathway (or glycolysis), as well as lipid metabolism (including both cholesterol and fatty acid metabolism) may impact some innate immune functions of PDC or may be involved in these functions after Toll-like receptor (TLR) 7/9 triggering. The kinetics of glycolysis after TLR7/9 triggering may differ between human and murine PDC. In mouse PDC, metabolism changes promoted by TLR7/9 activation may depend on an autocrine/paracrine loop, implicating type I IFN and its receptor IFNAR. This could explain a delayed glycolysis in mouse PDC. Moreover, PDC functions can be modulated by the metabolism of cholesterol and fatty acids. This may occur via the production of lipid ligands that activate nuclear receptors (e.g., liver X receptor [LXR]) in PDC or through limiting intracellular cholesterol pool size (by statin or LXR agonist treatment) in these cells. Finally, lipid-activated nuclear receptors (i.e., LXR or peroxisome proliferator activated receptor) may also directly interact with pro-inflammatory transcription factors, such as NF-κB. Here, we discuss how glycolysis and lipid metabolism may modulate PDC functions and how this may be harnessed in pathological situations where PDC play a detrimental role.
Collapse
Affiliation(s)
- Philippe Saas
- EFS Bourgogne Franche-Comté, Université Bourgogne Franche-Comté, Inserm, UMR1098, Besançon, F-25000, France
| | - Alexis Varin
- EFS Bourgogne Franche-Comté, Université Bourgogne Franche-Comté, Inserm, UMR1098, Besançon, F-25000, France
| | - Sylvain Perruche
- EFS Bourgogne Franche-Comté, Université Bourgogne Franche-Comté, Inserm, UMR1098, Besançon, F-25000, France
| | - Adam Ceroi
- EFS Bourgogne Franche-Comté, Université Bourgogne Franche-Comté, Inserm, UMR1098, Besançon, F-25000, France.,The Center for Cell Clearance, University of Virginia, Charlottesville, VA, 22903, USA
| |
Collapse
|
39
|
Saas P, Varin A, Perruche S, Ceroi A. Recent insights into the implications of metabolism in plasmacytoid dendritic cell innate functions: Potential ways to control these functions. F1000Res 2017; 6:456. [DOI: 10.12688/f1000research.11332.1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/07/2017] [Indexed: 12/12/2022] Open
Abstract
There are more and more data concerning the role of cellular metabolism in innate immune cells, such as macrophages or conventional dendritic cells. However, few data are available currently concerning plasmacytoid dendritic cells (PDC), another type of innate immune cells. These cells are the main type I interferon (IFN) producing cells, but they also secrete other pro-inflammatory cytokines (e.g., tumor necrosis factor or interleukin [IL]-6) or immunomodulatory factors (e.g., IL-10 or transforming growth factor-β). Through these functions, PDC participate in antimicrobial responses or maintenance of immune tolerance, and have been implicated in the pathophysiology of several autoimmune diseases. Recent data support the idea that the glycolytic pathway (or glycolysis), as well as lipid metabolism (including both cholesterol and fatty acid metabolism) may impact some innate immune functions of PDC or may be involved in these functions after Toll-like receptor (TLR) 7/9 triggering. Some differences may be related to the origin of PDC (human versus mouse PDC or blood-sorted versus FLT3 ligand stimulated-bone marrow-sorted PDC). The kinetics of glycolysis may differ between human and murine PDC. In mouse PDC, metabolism changes promoted by TLR7/9 activation may depend on an autocrine/paracrine loop, implicating type I IFN and its receptor IFNAR, explaining a delayed glycolysis. Moreover, PDC functions can be modulated by the metabolism of cholesterol and fatty acids. This may occur via the production of lipid ligands that activate nuclear receptors (e.g., liver X receptor [LXR]) in PDC or through limiting intracellular cholesterol pool size (by statins or LXR agonists) in these cells. Finally, lipid-activated nuclear receptors (i.e., LXR or peroxisome proliferator activated receptor) may also directly interact with pro-inflammatory transcription factors, such as NF-κB. Here, we discuss how glycolysis and lipid metabolism may modulate PDC functions and how this may be harnessed in pathological situations where PDC play a detrimental role.
Collapse
|
40
|
Schaheen B, Downs EA, Serbulea V, Almenara CCP, Spinosa M, Su G, Zhao Y, Srikakulapu P, Butts C, McNamara CA, Leitinger N, Upchurch GR, Meher AK, Ailawadi G. B-Cell Depletion Promotes Aortic Infiltration of Immunosuppressive Cells and Is Protective of Experimental Aortic Aneurysm. Arterioscler Thromb Vasc Biol 2016; 36:2191-2202. [PMID: 27634836 DOI: 10.1161/atvbaha.116.307559] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 09/02/2016] [Indexed: 01/09/2023]
Abstract
OBJECTIVE B-cell depletion therapy is widely used for treatment of cancers and autoimmune diseases. B cells are abundant in abdominal aortic aneurysms (AAA); however, it is unknown whether B-cell depletion therapy affects AAA growth. Using experimental models of murine AAA, we aim to examine the effect of B-cell depletion on AAA formation. APPROACH AND RESULTS Wild-type or apolipoprotein E-knockout mice were treated with mouse monoclonal anti-CD20 or control antibodies and subjected to an elastase perfusion or angiotensin II infusion model to induce AAA, respectively. Anti-CD20 antibody treatment significantly depleted B1 and B2 cells, and strikingly suppressed AAA growth in both models. B-cell depletion resulted in lower circulating IgM levels, but did not affect the levels of IgG or cytokine/chemokine levels. Although the total number of leukocyte remained unchanged in elastase-perfused aortas after anti-CD20 antibody treatment, the number of B-cell subtypes was significantly lower. Interestingly, plasmacytoid dendritic cells expressing the immunomodulatory enzyme indole 2,3-dioxygenase were detected in the aortas of B-cell-depleted mice. In accordance with an increase in indole 2,3-dioxygenase+ plasmacytoid dendritic cells, the number of regulatory T cells was higher, whereas the expression of proinflammatory genes was lower in aortas of B-cell-depleted mice. In a coculture model, the presence of B cells significantly lowered the number of indole 2,3-dioxygenase+ plasmacytoid dendritic cells without affecting total plasmacytoid dendritic cell number. CONCLUSIONS The present results demonstrate that B-cell depletion protects mice from experimental AAA formation and promotes emergence of an immunosuppressive environment in aorta.
Collapse
Affiliation(s)
- Basil Schaheen
- From the Departments of Surgery (B.S., E.A.D., M.S., G.S., Y.Z., G.R.U., A.K.M., G.A.), Pharmacology (V.S., C.C.P.A., N.L., A.K.M.), and Robert M. Berne Cardiovascular Research Center (P.S., C.A.M.N.), University of Virginia, Charlottesville; Biogen Idec, Cambridge, MA (C.B.); Department of Molecular Physiology and Biological Physics (G.R.U.) and Biomedical Engineering (G.A.), University of Virginia, Charlottesville
| | - Emily A Downs
- From the Departments of Surgery (B.S., E.A.D., M.S., G.S., Y.Z., G.R.U., A.K.M., G.A.), Pharmacology (V.S., C.C.P.A., N.L., A.K.M.), and Robert M. Berne Cardiovascular Research Center (P.S., C.A.M.N.), University of Virginia, Charlottesville; Biogen Idec, Cambridge, MA (C.B.); Department of Molecular Physiology and Biological Physics (G.R.U.) and Biomedical Engineering (G.A.), University of Virginia, Charlottesville
| | - Vlad Serbulea
- From the Departments of Surgery (B.S., E.A.D., M.S., G.S., Y.Z., G.R.U., A.K.M., G.A.), Pharmacology (V.S., C.C.P.A., N.L., A.K.M.), and Robert M. Berne Cardiovascular Research Center (P.S., C.A.M.N.), University of Virginia, Charlottesville; Biogen Idec, Cambridge, MA (C.B.); Department of Molecular Physiology and Biological Physics (G.R.U.) and Biomedical Engineering (G.A.), University of Virginia, Charlottesville
| | - Camila C P Almenara
- From the Departments of Surgery (B.S., E.A.D., M.S., G.S., Y.Z., G.R.U., A.K.M., G.A.), Pharmacology (V.S., C.C.P.A., N.L., A.K.M.), and Robert M. Berne Cardiovascular Research Center (P.S., C.A.M.N.), University of Virginia, Charlottesville; Biogen Idec, Cambridge, MA (C.B.); Department of Molecular Physiology and Biological Physics (G.R.U.) and Biomedical Engineering (G.A.), University of Virginia, Charlottesville
| | - Michael Spinosa
- From the Departments of Surgery (B.S., E.A.D., M.S., G.S., Y.Z., G.R.U., A.K.M., G.A.), Pharmacology (V.S., C.C.P.A., N.L., A.K.M.), and Robert M. Berne Cardiovascular Research Center (P.S., C.A.M.N.), University of Virginia, Charlottesville; Biogen Idec, Cambridge, MA (C.B.); Department of Molecular Physiology and Biological Physics (G.R.U.) and Biomedical Engineering (G.A.), University of Virginia, Charlottesville
| | - Gang Su
- From the Departments of Surgery (B.S., E.A.D., M.S., G.S., Y.Z., G.R.U., A.K.M., G.A.), Pharmacology (V.S., C.C.P.A., N.L., A.K.M.), and Robert M. Berne Cardiovascular Research Center (P.S., C.A.M.N.), University of Virginia, Charlottesville; Biogen Idec, Cambridge, MA (C.B.); Department of Molecular Physiology and Biological Physics (G.R.U.) and Biomedical Engineering (G.A.), University of Virginia, Charlottesville
| | - Yunge Zhao
- From the Departments of Surgery (B.S., E.A.D., M.S., G.S., Y.Z., G.R.U., A.K.M., G.A.), Pharmacology (V.S., C.C.P.A., N.L., A.K.M.), and Robert M. Berne Cardiovascular Research Center (P.S., C.A.M.N.), University of Virginia, Charlottesville; Biogen Idec, Cambridge, MA (C.B.); Department of Molecular Physiology and Biological Physics (G.R.U.) and Biomedical Engineering (G.A.), University of Virginia, Charlottesville
| | - Prasad Srikakulapu
- From the Departments of Surgery (B.S., E.A.D., M.S., G.S., Y.Z., G.R.U., A.K.M., G.A.), Pharmacology (V.S., C.C.P.A., N.L., A.K.M.), and Robert M. Berne Cardiovascular Research Center (P.S., C.A.M.N.), University of Virginia, Charlottesville; Biogen Idec, Cambridge, MA (C.B.); Department of Molecular Physiology and Biological Physics (G.R.U.) and Biomedical Engineering (G.A.), University of Virginia, Charlottesville
| | - Cherié Butts
- From the Departments of Surgery (B.S., E.A.D., M.S., G.S., Y.Z., G.R.U., A.K.M., G.A.), Pharmacology (V.S., C.C.P.A., N.L., A.K.M.), and Robert M. Berne Cardiovascular Research Center (P.S., C.A.M.N.), University of Virginia, Charlottesville; Biogen Idec, Cambridge, MA (C.B.); Department of Molecular Physiology and Biological Physics (G.R.U.) and Biomedical Engineering (G.A.), University of Virginia, Charlottesville
| | - Coleen A McNamara
- From the Departments of Surgery (B.S., E.A.D., M.S., G.S., Y.Z., G.R.U., A.K.M., G.A.), Pharmacology (V.S., C.C.P.A., N.L., A.K.M.), and Robert M. Berne Cardiovascular Research Center (P.S., C.A.M.N.), University of Virginia, Charlottesville; Biogen Idec, Cambridge, MA (C.B.); Department of Molecular Physiology and Biological Physics (G.R.U.) and Biomedical Engineering (G.A.), University of Virginia, Charlottesville
| | - Norbert Leitinger
- From the Departments of Surgery (B.S., E.A.D., M.S., G.S., Y.Z., G.R.U., A.K.M., G.A.), Pharmacology (V.S., C.C.P.A., N.L., A.K.M.), and Robert M. Berne Cardiovascular Research Center (P.S., C.A.M.N.), University of Virginia, Charlottesville; Biogen Idec, Cambridge, MA (C.B.); Department of Molecular Physiology and Biological Physics (G.R.U.) and Biomedical Engineering (G.A.), University of Virginia, Charlottesville
| | - Gilbert R Upchurch
- From the Departments of Surgery (B.S., E.A.D., M.S., G.S., Y.Z., G.R.U., A.K.M., G.A.), Pharmacology (V.S., C.C.P.A., N.L., A.K.M.), and Robert M. Berne Cardiovascular Research Center (P.S., C.A.M.N.), University of Virginia, Charlottesville; Biogen Idec, Cambridge, MA (C.B.); Department of Molecular Physiology and Biological Physics (G.R.U.) and Biomedical Engineering (G.A.), University of Virginia, Charlottesville
| | - Akshaya K Meher
- From the Departments of Surgery (B.S., E.A.D., M.S., G.S., Y.Z., G.R.U., A.K.M., G.A.), Pharmacology (V.S., C.C.P.A., N.L., A.K.M.), and Robert M. Berne Cardiovascular Research Center (P.S., C.A.M.N.), University of Virginia, Charlottesville; Biogen Idec, Cambridge, MA (C.B.); Department of Molecular Physiology and Biological Physics (G.R.U.) and Biomedical Engineering (G.A.), University of Virginia, Charlottesville.
| | - Gorav Ailawadi
- From the Departments of Surgery (B.S., E.A.D., M.S., G.S., Y.Z., G.R.U., A.K.M., G.A.), Pharmacology (V.S., C.C.P.A., N.L., A.K.M.), and Robert M. Berne Cardiovascular Research Center (P.S., C.A.M.N.), University of Virginia, Charlottesville; Biogen Idec, Cambridge, MA (C.B.); Department of Molecular Physiology and Biological Physics (G.R.U.) and Biomedical Engineering (G.A.), University of Virginia, Charlottesville
| |
Collapse
|
41
|
Mandl M, Drechsler M, Jansen Y, Neideck C, Noels H, Faussner A, Soehnlein O, Weber C, Döring Y. Evaluation of the BDCA2-DTR Transgenic Mouse Model in Chronic and Acute Inflammation. PLoS One 2015; 10:e0134176. [PMID: 26252890 PMCID: PMC4529211 DOI: 10.1371/journal.pone.0134176] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 07/06/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND AIMS Plasmacytoid dendritic cells (pDCs) are a small subset of dendritic cells and the main producers of type I interferons. Besides their contribution to tolerance, they are known to be involved in autoimmune diseases and have recently been implicated in atherosclerosis. However, their precise involvement, particularly in advanced lesion development, remains elusive. Hence, we investigated the role of pDCs in atherogenesis vs atheroprogression by specifically depleting this cell population using the BDCA2-DTR mouse model bred to Apolipoprotein E (Apoe-/-) deficient mice. METHODS AND RESULTS Our results revealed that continuous diphtheria toxin-induced pDC depletion in Apoe-/- BDCA2-DTR mice receiving a high-fat diet (HFD) for 4 weeks did not alter lesion size or composition. Instead, these mice displayed increased B cell numbers and altered levels of inflammatory cytokines. Analysis of depletion efficiency showed that complete pDC depletion could only be sustained for one week and reoccurring pDCs sorted after 4 weeks did not express DTR anymore. Consequently, we analyzed lesion development in a model of partial carotid ligation, inducing established lesions after 5 weeks of HFD feeding, and only depleted pDCs during the last week of 5 weeks HFD feeding. Despite short-term, but efficient pDC depletion, we observed no differences in atherosclerotic lesion development, but changes in inflammatory cytokine titers. To assure the functionality of the BDCA2-DTR model in acute settings, we additionally examined the effect of pDC depletion in an indirect acute lung injury (iALI) model. This time, efficient pDC depletion resulted in a significantly reduced macrophage and neutrophil accumulation in the lung 12 hours after LPS challenge, underlining a pro-inflammatory role of pDCs in the innate immune response in iALI. CONCLUSION Taken together, the BDCA2-DTR mouse model only allows efficient pDC depletion for one week, which subsequently restricts its usability to more acute but not chronic inflammatory disease models.
Collapse
Affiliation(s)
- Manuela Mandl
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Maik Drechsler
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany; Academic Medical Center, Department of Pathology, Amsterdam University, Amsterdam, the Netherlands; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Yvonne Jansen
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Carlos Neideck
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Heidi Noels
- Institute for Molecular Cardiovascular Research, RWTH Aachen University Aachen, Aachen, Germany
| | - Alexander Faussner
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Oliver Soehnlein
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany; Academic Medical Center, Department of Pathology, Amsterdam University, Amsterdam, the Netherlands; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Yvonne Döring
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
42
|
Tran Janco JM, Lamichhane P, Karyampudi L, Knutson KL. Tumor-infiltrating dendritic cells in cancer pathogenesis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 194:2985-91. [PMID: 25795789 PMCID: PMC4369768 DOI: 10.4049/jimmunol.1403134] [Citation(s) in RCA: 336] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Dendritic cells (DCs) play a pivotal role in the tumor microenvironment, which is known to affect disease progression in many human malignancies. Infiltration by mature, active DCs into the tumors confers an increase in immune activation and recruitment of disease-fighting immune effector cells and pathways. DCs are the preferential target of infiltrating T cells. However, tumor cells have means of suppressing DC function or of altering the tumor microenvironment in such a way that immune-suppressive DCs are recruited. Advances in understanding these changes have led to promising developments in cancer-therapeutic strategies targeting tumor-infiltrating DCs to subdue their immunosuppressive functions and enhance their immune-stimulatory capacity.
Collapse
Affiliation(s)
| | - Purushottam Lamichhane
- Department of Immunology, Mayo Clinic, Rochester, MN 55906; and Cancer Vaccines and Immune Therapies Program, Vaccine and Gene Therapy Institute, Port St. Lucie, FL 34987
| | - Lavakumar Karyampudi
- Cancer Vaccines and Immune Therapies Program, Vaccine and Gene Therapy Institute, Port St. Lucie, FL 34987
| | - Keith L Knutson
- Department of Immunology, Mayo Clinic, Rochester, MN 55906; and Cancer Vaccines and Immune Therapies Program, Vaccine and Gene Therapy Institute, Port St. Lucie, FL 34987
| |
Collapse
|