51
|
Monteiro S, Pinho AG, Macieira M, Serre-Miranda C, Cibrão JR, Lima R, Soares-Cunha C, Vasconcelos NL, Lentilhas-Graça J, Duarte-Silva S, Miranda A, Correia-Neves M, Salgado AJ, Silva NA. Splenic sympathetic signaling contributes to acute neutrophil infiltration of the injured spinal cord. J Neuroinflammation 2020; 17:282. [PMID: 32967684 PMCID: PMC7513542 DOI: 10.1186/s12974-020-01945-8] [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: 04/14/2020] [Accepted: 08/26/2020] [Indexed: 02/06/2023] Open
Abstract
Background Alterations in the immune system are a complication of spinal cord injury (SCI) and have been linked to an excessive sympathetic outflow to lymphoid organs. Still unknown is whether these peripheral immune changes also contribute for the deleterious inflammatory response mounted at the injured spinal cord. Methods We analyzed different molecular outputs of the splenic sympathetic signaling for the first 24 h after a thoracic compression SCI. We also analyzed the effect of ablating the splenic sympathetic signaling to the innate immune and inflammatory response at the spleen and spinal cord 24 h after injury. Results We found that norepinephrine (NE) levels were already raised at this time-point. Low doses of NE stimulation of splenocytes in vitro mainly affected the neutrophils’ population promoting an increase in both frequency and numbers. Interestingly, the interruption of the sympathetic communication to the spleen, by ablating the splenic nerve, resulted in reduced frequencies and numbers of neutrophils both at the spleen and spinal cord 1 day post-injury. Conclusion Collectively, our data demonstrates that the splenic sympathetic signaling is involved in the infiltration of neutrophils after spinal cord injury. Our findings give new mechanistic insights into the dysfunctional regulation of the inflammatory response mounted at the injured spinal cord.
Collapse
Affiliation(s)
- Susana Monteiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Portugal
| | - Andreia G Pinho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Portugal
| | - Mara Macieira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Portugal
| | - Cláudia Serre-Miranda
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Portugal
| | - Jorge R Cibrão
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Portugal
| | - Rui Lima
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Portugal
| | - Carina Soares-Cunha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Portugal
| | - Natália L Vasconcelos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Portugal
| | - José Lentilhas-Graça
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Portugal
| | - Sara Duarte-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Portugal
| | - Alice Miranda
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Portugal
| | - Margarida Correia-Neves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Portugal
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Portugal
| | - Nuno A Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal. .,ICVS/3B's-PT Government Associate Laboratory, Braga, Portugal.
| |
Collapse
|
52
|
Chen H, Wang R, Xu F, Zang T, Ji M, Yin J, Chen J, Shen L, Ge J. Renal denervation mitigates atherosclerosis in ApoE-/- mice via the suppression of inflammation. Am J Transl Res 2020; 12:5362-5380. [PMID: 33042425 PMCID: PMC7540133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 08/07/2020] [Indexed: 06/11/2023]
Abstract
Atherosclerosis is a chronic pathological process characterized by the accumulation of inflammation. Overactivation of the sympathetic nervous system accelerates the progression of atherosclerosis. Renal denervation (RDN) reduces the activity of the sympathetic nerve system (SNS) by disrupting sympathetic nerves surrounding renal arteries. We sought to determine whether RDN could mitigate atherosclerosis through the suppression of inflammation. First, we investigated the correlation between plasma norepinephrine concentrations and circulatory inflammation in the progression of atherosclerosis. Then, forty ApoE-/- mice underwent renal denervation or a sham operation after 6 weeks or 12 weeks of feeding with a high-fat diet. The effects of RDN on atherosclerosis in mice were explored. In the development of atherosclerosis, positive correlations were found between SNS activation and the accumulation of circulatory myeloid cells and inflammatory cytokines. In the second part of the study, inhibition of the increase in plaque size was found in both RDN groups compared with that in the sham operation (SO) groups (P<0.05), and RDN also ameliorated inflammation in plaques. Furthermore, RDN attenuated the accumulation of circulating neutrophils and monocytes (P<0.05), which is associated with a significant reduction in levels of several circulating inflammatory cytokines related to hemopoiesis (P<0.05). Flow cytometry analysis revealed comparable levels of neutrophils and monocytes in the bone marrow between all four groups. However, RDN decreased the production and proportions of neutrophils and monocytes in the spleen and reduced splenic sympathetic activity (P<0.05). In summary, our study reveals a novel link between SNS activity and inflammation in atherosclerosis and identifies RDN as a potential anti-inflammatory therapeutic strategy for the treatment of atherosclerosis by restricting the production of splenic immune cells.
Collapse
Affiliation(s)
- Han Chen
- Department of Cardiology, Zhongshan Hospital, Fudan UniversityShanghai, China
- Shanghai Institute of Cardiovascular DiseasesShanghai, China
| | - Rui Wang
- Department of Cardiology, Zhongshan Hospital, Fudan UniversityShanghai, China
- Shanghai Institute of Cardiovascular DiseasesShanghai, China
| | - Fei Xu
- Department of Cardiology, Zhongshan Hospital, Fudan UniversityShanghai, China
- Shanghai Institute of Cardiovascular DiseasesShanghai, China
| | - Tongtong Zang
- Department of Cardiology, Zhongshan Hospital, Fudan UniversityShanghai, China
- Shanghai Institute of Cardiovascular DiseasesShanghai, China
| | - Meng Ji
- Department of Cardiology, Zhongshan Hospital, Fudan UniversityShanghai, China
- Shanghai Institute of Cardiovascular DiseasesShanghai, China
| | - Jiasheng Yin
- Department of Cardiology, Zhongshan Hospital, Fudan UniversityShanghai, China
- Shanghai Institute of Cardiovascular DiseasesShanghai, China
| | - Jiahui Chen
- Department of Cardiology, Zhongshan Hospital, Fudan UniversityShanghai, China
- Shanghai Institute of Cardiovascular DiseasesShanghai, China
| | - Li Shen
- Department of Cardiology, Zhongshan Hospital, Fudan UniversityShanghai, China
- Shanghai Institute of Cardiovascular DiseasesShanghai, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan UniversityShanghai, China
- Shanghai Institute of Cardiovascular DiseasesShanghai, China
| |
Collapse
|
53
|
Nevin JT, Moussa M, Corwin WL, Mandoiu II, Srivastava PK. Sympathetic nervous tone limits the development of myeloid-derived suppressor cells. Sci Immunol 2020; 5:5/51/eaay9368. [PMID: 32917793 DOI: 10.1126/sciimmunol.aay9368] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 07/29/2020] [Indexed: 12/17/2022]
Abstract
Sympathetic nerves that innervate lymphoid organs regulate immune development and function by releasing norepinephrine that is sensed by immune cells via their expression of adrenergic receptors. Here, we demonstrate that ablation of sympathetic nervous system (SNS) signaling suppresses tumor immunity, and we dissect the mechanism of such immune suppression. We report that disruption of the SNS in mice removes a critical α-adrenergic signal required for maturation of myeloid cells in normal and tumor-bearing mice. In tumor-bearing mice, disruption of the α-adrenergic signal leads to the accumulation of immature myeloid-derived suppressor cells (MDSCs) that suppress tumor immunity and promote tumor growth. Furthermore, we show that these SNS-responsive MDSCs drive expansion of regulatory T cells via secretion of the alarmin heterodimer S100A8/A9, thereby compounding their immunosuppressive activity. Our results describe a regulatory framework in which sympathetic tone controls the development of innate and adaptive immune cells and influences their activity in health and disease.
Collapse
Affiliation(s)
- James T Nevin
- Department of Immunology and Carole and Ray Neag Comprehensive Cancer Center, University of Connecticut School of Medicine, Farmington, CT, USA.
| | - Marmar Moussa
- Department of Computer Science and Engineering, University of Connecticut, Farmington, CT, USA
| | - William L Corwin
- Department of Immunology and Carole and Ray Neag Comprehensive Cancer Center, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Ion I Mandoiu
- Department of Computer Science and Engineering, University of Connecticut, Farmington, CT, USA
| | - Pramod K Srivastava
- Department of Immunology and Carole and Ray Neag Comprehensive Cancer Center, University of Connecticut School of Medicine, Farmington, CT, USA.
| |
Collapse
|
54
|
Abstract
Communication between the nervous and immune systems is required for the body to regulate physiological homeostasis. Beta-adrenergic receptors expressed on immune cells mediate the modulation of immune response by neural activity. Activation of beta-adrenergic signaling results in suppression of antitumor immune response and limits the efficacy of cancer immunotherapy. Beta-adrenergic signaling is also involved in regulation of hematopoietic reconstitution, which is critical to the graft-versus-tumor (GVT) effect and to graft-versus-host disease (GVHD) following allogeneic hematopoietic cell transplantation (HCT). In this review, the function of beta-adrenergic signaling in mediating tumor immunosuppression will be highlighted. We will also discuss the implication of targeting beta-adrenergic signaling to improve the efficacy of cancer immunotherapy including the GVT effect, and to diminish the adverse effects including GVHD.
Collapse
Affiliation(s)
- Wei Wang
- Department of Microbiology and Immunology, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland
| | - Xuefang Cao
- Department of Microbiology and Immunology, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland
| |
Collapse
|
55
|
Barman PK, Koh TJ. Macrophage Dysregulation and Impaired Skin Wound Healing in Diabetes. Front Cell Dev Biol 2020; 8:528. [PMID: 32671072 PMCID: PMC7333180 DOI: 10.3389/fcell.2020.00528] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 06/05/2020] [Indexed: 12/13/2022] Open
Abstract
Monocytes (Mo) and macrophages (Mϕ) play important roles in normal skin wound healing, and dysregulation of wound Mo/Mϕ leads to impaired wound healing in diabetes. Although skin wound Mϕ originate both from tissue resident Mϕ and infiltrating bone marrow-derived Mo, the latter play dominant roles during the inflammatory phase of wound repair. Increased production of bone marrow Mo caused by alterations of hematopoietic stem and progenitor cell (HSPC) niche and epigenetic modifications of HSPCs likely contributes to the enhanced number of wound Mϕ in diabetes. In addition, an impaired transition of diabetic wound Mϕ from “pro-inflammatory” to “pro-healing” phenotypes driven by the local wound environment as well as intrinsic changes in bone marrow Mo is also thought to be partly responsible for impaired diabetic wound healing. The current brief review describes the origin, heterogeneity and function of wound Mϕ during normal skin wound healing followed by discussion of how dysregulated wound Mϕ numbers and phenotype are associated with impaired diabetic wound healing. The review also highlights the possible links between altered bone marrow myelopoiesis and increased Mo production as well as extrinsic and intrinsic factors that drive wound macrophage dysregulation leading to impaired wound healing in diabetes.
Collapse
Affiliation(s)
- Pijus K Barman
- Department of Kinesiology and Nutrition, Center for Wound Healing and Tissue Regeneration, University of Illinois at Chicago, Chicago, IL, United States
| | - Timothy J Koh
- Department of Kinesiology and Nutrition, Center for Wound Healing and Tissue Regeneration, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
56
|
Mohammadpour H, Sarow JL, MacDonald CR, Chen GL, Qiu J, Sharma UC, Cao X, Herr MM, Hahn TE, Blazar BR, Repasky EA, McCarthy PL. β2-Adrenergic receptor activation on donor cells ameliorates acute GvHD. JCI Insight 2020; 5:137788. [PMID: 32437333 DOI: 10.1172/jci.insight.137788] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 05/13/2020] [Indexed: 12/12/2022] Open
Abstract
Acute graft versus host disease (aGvHD) remains a major impediment to successful allogeneic hematopoietic cell transplantation (allo-HCT). To solve this problem, a greater knowledge of factors that regulate the differentiation of donor T cells toward cytotoxic cells or Tregs is necessary. We report that the β2-adrenergic receptor (β2-AR) is critical for regulating this differentiation and that its manipulation can control aGvHD without impairing the graft-versus-tumor (GvT) effect. Donor T cell β2-AR expression and signaling is associated with decreased aGvHD when compared with recipients of β2-AR-/- donor T cells. We determined that β2-AR activation skewed CD4+ T cell differentiation in vitro and in vivo toward Tregs rather than the T helper 1 (Th1) phenotype. Treatment of allo-HCT recipients with a selective β2-agonist (bambuterol) ameliorated aGvHD severity. This was associated with increased Tregs, decreased cytotoxic T cells, and increased donor BM-derived myeloid-derived suppressor cells (MDSCs) in allogeneic and humanized xenogeneic aGvHD models. β2-AR signaling resulted in increased Treg generation through glycogen synthase kinase-3 activation. Bambuterol preserved the GvT effect by inducing NKG2D+ effector cells and central memory T cells. These data reveal how β-AR signaling can be targeted to ameliorate GvHD severity while preserving GvT effect.
Collapse
Affiliation(s)
| | | | | | - George L Chen
- Medicine, Transplant and Cellular Therapy Program, and
| | - Jingxin Qiu
- Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Umesh C Sharma
- Department of Medicine, Jacobs School of Medicine & Biomedical Sciences, Buffalo, New York, USA
| | - Xuefang Cao
- Department of Microbiology and Immunology, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland, USA
| | - Megan M Herr
- Medicine, Transplant and Cellular Therapy Program, and
| | | | - Bruce R Blazar
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | | | | |
Collapse
|
57
|
Wu C, Hua Q, Zheng L. Generation of Myeloid Cells in Cancer: The Spleen Matters. Front Immunol 2020; 11:1126. [PMID: 32582203 PMCID: PMC7291604 DOI: 10.3389/fimmu.2020.01126] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 05/07/2020] [Indexed: 02/06/2023] Open
Abstract
Myeloid cells are key components of the tumor microenvironment and critical regulators of disease progression. These innate immune cells are usually short-lived and require constant replenishment. Emerging evidence indicates that tumors alter the host hematopoietic system and induce the biased differentiation of myeloid cells to tip the balance of the systemic immune activities toward tumor-promoting functions. Altered myelopoiesis is not restricted to the bone marrow and also occurs in extramedullary organs. In this review, we outline the recent advances in the field of cancer-associated myelopoiesis, with a focus on the spleen, the major site of extramedullary hematopoiesis in the cancer setting. We discuss the functional specialization, distinct mechanisms, and clinical relevance of cancer-associated myeloid cell generation from early progenitors in the spleen and its potential as a novel therapeutic target.
Collapse
Affiliation(s)
- Chong Wu
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Qiaomin Hua
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Limin Zheng
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
58
|
Zhao J, Florentin J, Tai YY, Torrino S, Ohayon L, Brzoska T, Tang Y, Yang J, Negi V, Woodcock CSC, Risbano MG, Nouraie SM, Sundd P, Bertero T, Dutta P, Chan SY. Long Range Endocrine Delivery of Circulating miR-210 to Endothelium Promotes Pulmonary Hypertension. Circ Res 2020; 127:677-692. [PMID: 32493166 DOI: 10.1161/circresaha.119.316398] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
RATIONALE Unproven theories abound regarding the long-range uptake and endocrine activity of extracellular blood-borne microRNAs into tissue. In pulmonary hypertension (PH), microRNA-210 (miR-210) in pulmonary endothelial cells promotes disease, but its activity as an extracellular molecule is incompletely defined. OBJECTIVE We investigated whether chronic and endogenous endocrine delivery of extracellular miR-210 to pulmonary vascular endothelial cells promotes PH. METHODS AND RESULTS Using miR-210 replete (wild-type [WT]) and knockout mice, we tracked blood-borne miR-210 using bone marrow transplantation and parabiosis (conjoining of circulatory systems). With bone marrow transplantation, circulating miR-210 was derived predominantly from bone marrow. Via parabiosis during chronic hypoxia to induce miR-210 production and PH, miR-210 was undetectable in knockout-knockout mice pairs. However, in plasma and lung endothelium, but not smooth muscle or adventitia, miR-210 was observed in knockout mice of WT-knockout pairs. This was accompanied by downregulation of miR-210 targets ISCU (iron-sulfur assembly proteins)1/2 and COX10 (cytochrome c oxidase assembly protein-10), indicating endothelial import of functional miR-210. Via hemodynamic and histological indices, knockout-knockout pairs were protected from PH, whereas knockout mice in WT-knockout pairs developed PH. In particular, pulmonary vascular engraftment of miR-210-positive interstitial lung macrophages was observed in knockout mice of WT-knockout pairs. To address whether engrafted miR-210-positive myeloid or lymphoid cells contribute to paracrine miR-210 delivery, we studied miR-210 knockout mice parabiosed with miR-210 WT; Cx3cr1 knockout mice (deficient in myeloid recruitment) or miR-210 WT; Rag1 knockout mice (deficient in lymphocytes). In both pairs, miR-210 knockout mice still displayed miR-210 delivery and PH, thus demonstrating a pathogenic endocrine delivery of extracellular miR-210. CONCLUSIONS Endogenous blood-borne transport of miR-210 into pulmonary vascular endothelial cells promotes PH, offering fundamental insight into the systemic physiology of microRNA activity. These results also describe a platform for RNA-mediated crosstalk in PH, providing an impetus for developing blood-based miR-210 technologies for diagnosis and therapy in this disease.
Collapse
Affiliation(s)
- Jingsi Zhao
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute (J.Z., J.F., Y.-Y.T., L.O., T. Brzoska, Y.T., J.Y., V.N., C.-S.C.W., M.G.R., S.M.N., P.S., P.D., S.Y.C.), University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
| | - Jonathan Florentin
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute (J.Z., J.F., Y.-Y.T., L.O., T. Brzoska, Y.T., J.Y., V.N., C.-S.C.W., M.G.R., S.M.N., P.S., P.D., S.Y.C.), University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
| | - Yi-Yin Tai
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute (J.Z., J.F., Y.-Y.T., L.O., T. Brzoska, Y.T., J.Y., V.N., C.-S.C.W., M.G.R., S.M.N., P.S., P.D., S.Y.C.), University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
| | - Stéphanie Torrino
- Université Côte d'Azur, CNRS, IPMC, Valbonne, France (S.T., T. Bertero)
| | - Lee Ohayon
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute (J.Z., J.F., Y.-Y.T., L.O., T. Brzoska, Y.T., J.Y., V.N., C.-S.C.W., M.G.R., S.M.N., P.S., P.D., S.Y.C.), University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
| | - Tomasz Brzoska
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute (J.Z., J.F., Y.-Y.T., L.O., T. Brzoska, Y.T., J.Y., V.N., C.-S.C.W., M.G.R., S.M.N., P.S., P.D., S.Y.C.), University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
| | - Ying Tang
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute (J.Z., J.F., Y.-Y.T., L.O., T. Brzoska, Y.T., J.Y., V.N., C.-S.C.W., M.G.R., S.M.N., P.S., P.D., S.Y.C.), University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
| | - Jimin Yang
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute (J.Z., J.F., Y.-Y.T., L.O., T. Brzoska, Y.T., J.Y., V.N., C.-S.C.W., M.G.R., S.M.N., P.S., P.D., S.Y.C.), University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
| | - Vinny Negi
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute (J.Z., J.F., Y.-Y.T., L.O., T. Brzoska, Y.T., J.Y., V.N., C.-S.C.W., M.G.R., S.M.N., P.S., P.D., S.Y.C.), University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
| | - Chen-Shan Chen Woodcock
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute (J.Z., J.F., Y.-Y.T., L.O., T. Brzoska, Y.T., J.Y., V.N., C.-S.C.W., M.G.R., S.M.N., P.S., P.D., S.Y.C.), University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
| | - Michael G Risbano
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute (J.Z., J.F., Y.-Y.T., L.O., T. Brzoska, Y.T., J.Y., V.N., C.-S.C.W., M.G.R., S.M.N., P.S., P.D., S.Y.C.), University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA.,Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine (M.G.R., S.M.N., P.S.), University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
| | - Seyed Mehdi Nouraie
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute (J.Z., J.F., Y.-Y.T., L.O., T. Brzoska, Y.T., J.Y., V.N., C.-S.C.W., M.G.R., S.M.N., P.S., P.D., S.Y.C.), University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA.,Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine (M.G.R., S.M.N., P.S.), University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
| | - Prithu Sundd
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute (J.Z., J.F., Y.-Y.T., L.O., T. Brzoska, Y.T., J.Y., V.N., C.-S.C.W., M.G.R., S.M.N., P.S., P.D., S.Y.C.), University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA.,Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine (M.G.R., S.M.N., P.S.), University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
| | - Thomas Bertero
- Université Côte d'Azur, CNRS, IPMC, Valbonne, France (S.T., T. Bertero)
| | - Partha Dutta
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute (J.Z., J.F., Y.-Y.T., L.O., T. Brzoska, Y.T., J.Y., V.N., C.-S.C.W., M.G.R., S.M.N., P.S., P.D., S.Y.C.), University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA.,Division of Cardiology, Department of Medicine (P.D., S.Y.C.), University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
| | - Stephen Y Chan
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute (J.Z., J.F., Y.-Y.T., L.O., T. Brzoska, Y.T., J.Y., V.N., C.-S.C.W., M.G.R., S.M.N., P.S., P.D., S.Y.C.), University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA.,Division of Cardiology, Department of Medicine (P.D., S.Y.C.), University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
| |
Collapse
|
59
|
Carnevale D, Lembo G. Neuroimmune interactions in cardiovascular diseases. Cardiovasc Res 2020; 117:402-410. [PMID: 32462184 DOI: 10.1093/cvr/cvaa151] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 04/27/2020] [Accepted: 05/22/2020] [Indexed: 02/07/2023] Open
Abstract
Our body is continuously in contact with external stimuli that need a fine integration with the internal milieu in order to maintain the homoeostasis. Similarly, perturbations of the internal environment are responsible for the alterations of the physiological mechanisms regulating our main functions. The nervous system and the immune system represent the main interfaces between the internal and the external environment. In carrying out these functions, they share many similarities, being able to recognize, integrate, and organize responses to a wide variety of stimuli, with the final aim to re-establish the homoeostasis. The autonomic nervous system, which collectively refers to the ensemble of afferent and efferent neurons that wire the central nervous system with visceral effectors throughout the body, is the prototype system controlling the homoeostasis through reflex arches. On the other hand, immune cells continuously patrol our body against external enemies and internal perturbations, organizing acute responses and forming memory for future encounters. Interesting to notice, the integration of the two systems provides a further unique opportunity for fine tuning of our body's homoeostasis. In fact, the autonomic nervous system guides the development of lymphoid and myeloid organs, as well as the deployment of immune cells towards peripheral tissues where they can affect and control several physiological functions. In turn, every specific immune cell type can contribute to regulate neural circuits involved in cardiovascular function, metabolism, and inflammation. Here, we review current understanding of the cross-regulation between these systems in cardiovascular diseases.
Collapse
Affiliation(s)
- Daniela Carnevale
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, Via dell'Elettronica, 86077 Pozzilli IS, Italy.,Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| | - Giuseppe Lembo
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, Via dell'Elettronica, 86077 Pozzilli IS, Italy.,Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| |
Collapse
|
60
|
Jiao X, Lu D, Pei X, Qi D, Huang S, Song Z, Gu J, Li Z. Type 1 diabetes mellitus impairs diurnal oscillations in murine extraorbital lacrimal glands. Ocul Surf 2020; 18:438-452. [PMID: 32360784 DOI: 10.1016/j.jtos.2020.04.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 04/20/2020] [Accepted: 04/23/2020] [Indexed: 01/09/2023]
Abstract
PURPOSE People with diabetes are at high risk of lacrimal gland dysfunction, but the underlying mechanism is not well understood. In this study, we determined how type 1 diabetes mellitus (T1DM) influences circadian homeostasis of the murine extraorbital lacrimal glands (ELGs). METHODS A T1DM animal model was established by systemic streptozotocin injection in C57BL/6J mice. After 5 weeks, ELGs were collected at 3-h intervals over a 24-h circadian cycle. Total extracted RNA was subjected to high-throughput RNA sequencing, and rhythmic transcriptional data were evaluated using the Jonckheere-Terpstra-Kendall algorithm, Kyoto Encyclopedia of Genes and Genomes pathway analysis, Phase Set Enrichment Analysis, and time series cluster analysis to determine the phase, rhythmicity, and unique signature of the transcripts over temporally coordinated expression. Additionally, mass, cell size, histology, and tear secretion of the ELGs were evaluated. RESULTS T1DM globally altered the composition of the ELG transcriptome. Specifically, T1DM significantly reprogrammed the circadian transcriptomic profiles of normal ELGs and reorganized core clock machinery. Unique temporal and clustering enrichment pathways were also rewired by T1DM. Finally, normal daily rhythms of mass, cell size, and tear secretion of mouse ELGs were significantly impaired by streptozotocin-induced diabetes. CONCLUSIONS T1DM significantly reprograms the diurnal oscillations of the lacrimal glands and impairs their structure and tear secretion. This information may reveal potential targets for improving lacrimal gland dysfunction in patients with diabetes.
Collapse
Affiliation(s)
- Xinwei Jiao
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Dingli Lu
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Xiaoting Pei
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Di Qi
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Shenzhen Huang
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Zongming Song
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Jianqin Gu
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Zhijie Li
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China.
| |
Collapse
|
61
|
An in vivo brain-bacteria interface: the developing brain as a key regulator of innate immunity. NPJ Regen Med 2020; 5:2. [PMID: 32047653 PMCID: PMC7000827 DOI: 10.1038/s41536-020-0087-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 12/20/2019] [Indexed: 01/11/2023] Open
Abstract
Infections have numerous effects on the brain. However, possible roles of the brain in protecting against infection, and the developmental origin and role of brain signaling in immune response, are largely unknown. We exploited a unique Xenopus embryonic model to reveal control of innate immune response to pathogenic E. coli by the developing brain. Using survival assays, morphological analysis of innate immune cells and apoptosis, and RNA-seq, we analyzed combinations of infection, brain removal, and tail-regenerative response. Without a brain, survival of embryos injected with bacteria decreased significantly. The protective effect of the developing brain was mediated by decrease of the infection-induced damage and of apoptosis, and increase of macrophage migration, as well as suppression of the transcriptional consequences of the infection, all of which decrease susceptibility to pathogen. Functional and pharmacological assays implicated dopamine signaling in the bacteria–brain–immune crosstalk. Our data establish a model that reveals the very early brain to be a central player in innate immunity, identify the developmental origins of brain–immune interactions, and suggest several targets for immune therapies.
Collapse
|
62
|
Sympathetic nervous activity and hemoglobin levels in de novo Parkinson’s disease. Clin Auton Res 2020; 30:273-278. [DOI: 10.1007/s10286-020-00668-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 01/17/2020] [Indexed: 12/13/2022]
|
63
|
Christ A, Lauterbach M, Latz E. Western Diet and the Immune System: An Inflammatory Connection. Immunity 2020; 51:794-811. [PMID: 31747581 DOI: 10.1016/j.immuni.2019.09.020] [Citation(s) in RCA: 417] [Impact Index Per Article: 104.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 07/24/2019] [Accepted: 09/24/2019] [Indexed: 02/06/2023]
Abstract
The consumption of Western-type calorically rich diets combined with chronic overnutrition and a sedentary lifestyle in Western societies evokes a state of chronic metabolic inflammation, termed metaflammation. Metaflammation contributes to the development of many prevalent non-communicable diseases (NCDs), and these lifestyle-associated pathologies represent a rising public health problem with global epidemic dimensions. A better understanding of how modern lifestyle and Western diet (WD) activate immune cells is essential for the development of efficient preventive and therapeutic strategies for common NCDs. Here, we review the current mechanistic understanding of how the Western lifestyle can induce metaflammation, and we discuss how this knowledge can be translated to protect the public from the health burden associated with their selected lifestyle.
Collapse
Affiliation(s)
- Anette Christ
- Institute of Innate Immunity, University Hospital Bonn, University of Bonn, Bonn 53127, Germany; Department of Infectious Diseases & Immunology, UMass Medical School, Worcester, MA 01605, USA
| | - Mario Lauterbach
- Institute of Innate Immunity, University Hospital Bonn, University of Bonn, Bonn 53127, Germany
| | - Eicke Latz
- Institute of Innate Immunity, University Hospital Bonn, University of Bonn, Bonn 53127, Germany; Department of Infectious Diseases & Immunology, UMass Medical School, Worcester, MA 01605, USA; Center of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim 7491, Norway; German Center for Neurodegenerative Diseases (DZNE), Bonn 53127, Germany.
| |
Collapse
|
64
|
Renal denervation restrains the inflammatory response in myocardial ischemia-reperfusion injury. Basic Res Cardiol 2020; 115:15. [PMID: 31932910 DOI: 10.1007/s00395-020-0776-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 01/02/2020] [Indexed: 12/19/2022]
Abstract
Myocardial ischemia-reperfusion (I/R) injury leads to intensive sympathetic nervous system (SNS) activation and inflammatory reactions. Whether renal sympathetic denervation (RDN) could be a new therapeutic strategy to modulate I/R inflammation and reduce infarct size after myocardial I/R injury needs to be explored. First, we investigated the correlation between plasma norepinephrine concentrations and circulating myeloid cell numbers in patients with acute myocardial infarction. And then, C57BL/6 mice underwent a "two-hit" operation, with 10% phenol applied to bilateral renal nerves to abrogate sympathoexcitation, and a 45-min ligation of the left coronary artery to induce myocardial I/R injury. The effects of RDN on the mobilization of immune cells in mice following myocardial I/R injury were explored. We observed a strong association between SNS overactivation and myeloid cell excessive accumulation in patients. In animal experiments, there was a significant reduction in infarct size per area at risk in the denervated-I/R group when compared to that of the innervated-I/R group (39.2% versus 49.8%; p < 0.005), and RDN also improved the left ventricular ejection fraction by 20% after 1 week. Furthermore, the denervated-I/R group showed a decrease in the number of neutrophils and macrophages in the blood and the myocardium as reflected by immunohistochemical staining and flow cytometry analysis (p < 0.05); the decrease was associated with a significant reduction in the circulating production of IL-1, IL-6 and TNF-α (p < 0.05). In summary, our study reveals a novel link between the SNS activity and inflammatory response undergoing myocardium I/R injury and identifies RDN as a potential therapeutic strategy against myocardium I/R injury via preserving the spleen immune cells mobilization.
Collapse
|
65
|
Straub RH, Dufner B, Rauch L. Proinflammatory α-Adrenergic Neuronal Regulation of Splenic IFN-γ, IL-6, and TGF-β of Mice from Day 15 onwards in Arthritis. Neuroimmunomodulation 2020; 27:58-68. [PMID: 32610310 PMCID: PMC7446300 DOI: 10.1159/000508109] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 04/19/2020] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION In arthritic mice, a sympathetic influence is proinflammatory from the time point of immunization until the onset of disease (days 0-32), but reasons are unknown. Disruption of the major anti-inflammatory pathway through Gαs-coupled receptors probably play a role. For example, noradrenaline cannot operate via anti-inflammatory β2-adrenoceptors but through proinflammatory α1/2-ad-renoceptors. This might happen, first, through a loss of sympathetic nerve fibers in inflamed tissue with low neurotransmitter levels (noradrenaline only binds to high-affinity α-adrenoceptors) and, second, through an alteration in G-protein receptor coupling with a predominance of α-adrenergic signaling. We hypothesized that both mechanisms play a role in the course of collagen type II-induced arthritis (CIA) in the spleen in mice. METHODS In CIA mice, nerve fiber density in the spleen was quantified by immunohistochemistry techniques. The functional impact of sympathetic nerve fibers in the spleen was studied by a micro-superfusion technique of spleen slices with a focus on the secretion of IFN-γ and IL-6 (proinflammatory) and TGF-β (anti-inflammatory). RESULTS During CIA, sympathetic nerve fibers get increasingly lost from day14 until day 55 after immunization. The influence of electrically released noradrenaline diminishes in the course of arthritis. At all investigated time points (days 14, 32, and 55), only proinflammatory neuronal α-adrenergic effects on cytokine secretion were demonstrated (i.e., stimulation of IFN-γ and IL-6 and inhibition of TGF-β). CONCLUSION Sympathetic nerve fibers are rapidly lost in the spleen, and only proinflammatory α-adrenergic neuronal regulation of cytokine secretion takes place throughout the course of arthritis. These results support a predominance of a proinflammatory α-adrenergic sympathetic influence in arthritis.
Collapse
Affiliation(s)
- Rainer H Straub
- Laboratory of Experimental Rheumatology and Neuroendocrine Immunology, Department of Internal Medicine, University Hospital, Regensburg, Germany,
| | - Bianca Dufner
- Laboratory of Experimental Rheumatology and Neuroendocrine Immunology, Department of Internal Medicine, University Hospital, Regensburg, Germany
| | - Luise Rauch
- Laboratory of Experimental Rheumatology and Neuroendocrine Immunology, Department of Internal Medicine, University Hospital, Regensburg, Germany
| |
Collapse
|
66
|
Mohammadpour H, MacDonald CR, Qiao G, Chen M, Dong B, Hylander BL, McCarthy PL, Abrams SI, Repasky EA. β2 adrenergic receptor-mediated signaling regulates the immunosuppressive potential of myeloid-derived suppressor cells. J Clin Invest 2019; 129:5537-5552. [PMID: 31566578 PMCID: PMC6877316 DOI: 10.1172/jci129502] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 09/18/2019] [Indexed: 12/11/2022] Open
Abstract
Catecholamines released by sympathetic nerves can activate adrenergic receptors present on nearly every cell type, including myeloid-derived suppressor cells (MDSCs). Using in vitro systems, murine tumor models in wild-type and genetically modified (β2-AR-/-) mice, and adoptive transfer approaches, we found that the degree of β2-AR signaling significantly influences MDSC frequency and survival in tumors and other tissues. It also modulates their expression of immunosuppressive molecules such as arginase-I and PD-L1 and alters their ability to suppress the proliferation of T cells. The regulatory functions of β2-AR signaling in MDSCs were also found to be dependent upon STAT3 phosphorylation. Moreover, we observed that the β2-AR-mediated increase in MDSC survival is dependent upon Fas-FasL interactions, and this is consistent with gene expression analyses, which reveal a greater expression of apoptosis-related genes in β2-AR-/- MDSCs. Our data reveal the potential of β2-AR signaling to increase the generation of MDSCs from both murine and human peripheral blood cells and that the immunosuppressive function of MDSCs can be mitigated by treatment with β-AR antagonists, or enhanced by β-AR agonists. This strongly supports the possibility that reducing stress-induced activation of β2-ARs could help to overcome immune suppression and enhance the efficacy of immunotherapy and other cancer therapies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Philip L. McCarthy
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | | | | |
Collapse
|
67
|
Kanashiro A, Hiroki CH, da Fonseca DM, Birbrair A, Ferreira RG, Bassi GS, Fonseca MD, Kusuda R, Cebinelli GCM, da Silva KP, Wanderley CW, Menezes GB, Alves-Fiho JC, Oliveira AG, Cunha TM, Pupo AS, Ulloa L, Cunha FQ. The role of neutrophils in neuro-immune modulation. Pharmacol Res 2019; 151:104580. [PMID: 31786317 DOI: 10.1016/j.phrs.2019.104580] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/07/2019] [Accepted: 11/27/2019] [Indexed: 01/10/2023]
Abstract
Neutrophils are peripheral immune cells that represent the first recruited innate immune defense against infections and tissue injury. However, these cells can also induce overzealous responses and cause tissue damage. Although the role of neutrophils activating the immune system is well established, only recently their critical implications in neuro-immune interactions are becoming more relevant. Here, we review several aspects of neutrophils in the bidirectional regulation between the nervous and immune systems. First, the role of neutrophils as a diffuse source of acetylcholine and catecholamines is controversial as well as the effects of these neurotransmitters in neutrophil's functions. Second, neutrophils contribute for the activation and sensitization of sensory neurons, and thereby, in events of nociception and pain. In addition, nociceptor activation promotes an axon reflex triggering a local release of neural mediators and provoking neutrophil activation. Third, the recruitment of neutrophils in inflammatory responses in the nervous system suggests these immune cells as innovative targets in the treatment of central infectious, neurological and neurodegenerative disorders. Multidisciplinary studies involving immunologists and neuroscientists are required to define the role of the neurons-neutrophils communication in the pathophysiology of infectious, inflammatory, and neurological disorders.
Collapse
Affiliation(s)
- Alexandre Kanashiro
- Department of Neurosciences and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil; Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil.
| | - Carlos Hiroji Hiroki
- Department of Immunology and Biochemistry, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Denise Morais da Fonseca
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Alexander Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Raphael Gomes Ferreira
- Araguaína Medical School, Federal University of Tocantins, Avenida Paraguai s/n, 77824-838, Araguaína, TO, Brazil
| | - Gabriel Shimizu Bassi
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University, Durham, NC, 27710, USA
| | - Mirian D Fonseca
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Ricardo Kusuda
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | | | - Katiussia Pinho da Silva
- Department of Pharmacology, Instituto de Biociências, Universidade Estadual Paulista, Botucatu, SP, Brazil
| | - Carlos Wagner Wanderley
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | | | - José Carlos Alves-Fiho
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - André Gustavo Oliveira
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Thiago M Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - André Sampaio Pupo
- Department of Pharmacology, Instituto de Biociências, Universidade Estadual Paulista, Botucatu, SP, Brazil
| | - Luis Ulloa
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University, Durham, NC, 27710, USA.
| | - Fernando Queiroz Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
68
|
Tanaka S, Okusa MD. Crosstalk between the nervous system and the kidney. Kidney Int 2019; 97:466-476. [PMID: 32001065 DOI: 10.1016/j.kint.2019.10.032] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/25/2019] [Accepted: 10/28/2019] [Indexed: 12/20/2022]
Abstract
Under physiological states, the nervous system and the kidneys communicate with each other to maintain normal body homeostasis. However, pathological states disrupt this interaction as seen in hypertension, and kidney damage can cause impaired renorenal reflex and sodium handling. In acute kidney injury (AKI) and chronic kidney disease (CKD), damaged kidneys can have a detrimental effect on the central nervous system. CKD is an independent risk factor for cerebrovascular disease and cognitive impairment, and many factors, including retention of uremic toxins and phosphate, have been proposed as CKD-specific factors responsible for structural and functional cerebral changes in patients with CKD. However, more studies are needed to determine the precise pathogenesis. Epidemiological studies have shown that AKI is associated with a subsequent risk for developing stroke and dementia. However, recent animal studies have shown that the renal nerve contributes to kidney inflammation and fibrosis, whereas activation of the cholinergic anti-inflammatory pathway, which involves the vagus nerve, the splenic nerve, and immune cells in the spleen, has a significant renoprotective effect. Therefore, elucidating mechanisms of communication between the nervous system and the kidney enables us not only to develop new strategies to ameliorate neurological conditions associated with kidney disease but also to design safe and effective clinical interventions for kidney disease, using the neural and neuroimmune control of kidney injury and disease.
Collapse
Affiliation(s)
- Shinji Tanaka
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Mark D Okusa
- Division of Nephrology and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, Virginia, USA.
| |
Collapse
|
69
|
Wu H, Song Y, Li J, Lei X, Zhang S, Gao Y, Cheng P, Liu B, Miao S, Bi L, Yang L, Pei G. Blockade of adrenergic β-receptor activation through local delivery of propranolol from a 3D collagen/polyvinyl alcohol/hydroxyapatite scaffold promotes bone repair in vivo. Cell Prolif 2019; 53:e12725. [PMID: 31746058 PMCID: PMC6985692 DOI: 10.1111/cpr.12725] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 10/19/2019] [Accepted: 10/31/2019] [Indexed: 12/14/2022] Open
Abstract
Objectives Activation of the sympathetic system and adrenergic β‐receptors following traumatic bone defects negatively impairs bone regeneration. Whether preventing β‐receptor activation could potentially improve bone defect repair is unknown. In this study, we investigated the effect of systematic administration and local delivery of propranolol through composite scaffolds on bone healing. Materials and methods Collagen/PVA/propranolol/hydroxyapatite(CPPH)composite scaffolds were fabricated with 3D printing technique and characterized by scanning electron microscope (SEM). Micro‐CT analysis and bone formation histology were performed to detect new bone formation. Osteogenic differentiation of bone marrow stromal cells (BMSCs) and osteoclastogenesis of bone marrow monocytes cultured with scaffolds extract were performed for further verification. Results Intraperitoneal injection of propranolol did not significantly improve bone repair, as indicated by micro‐CT analysis and bone formation histology. However, CPPH scaffolds exhibited sustained release of propranolol in vitro and significantly enhanced bone regeneration compared with vehicle collagen/PVA/hydroxyapatite (CPH) scaffolds in vivo. Moreover, in vitro experiments indicated the scaffolds containing propranolol promoted the osteogenic differentiation and migration of rat BMSCs and inhibited osteoclastogenesis by preventing β‐receptor activation. Conclusions This study demonstrates that local adrenergic β‐receptor blockade can effectively enhance the treatment of bone defects by stimulating osteogenic differentiation, inhibiting osteoclastogenesis and enhancing BMSCs migration.
Collapse
Affiliation(s)
- Hao Wu
- Department of Orthopaedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yue Song
- Department of Orthopaedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Junqin Li
- Department of Orthopaedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xing Lei
- Department of Orthopaedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China.,Department of Orthopedic Surgery, Linyi People's Hospital, Linyi, China
| | - Shuaishuai Zhang
- Department of Orthopaedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yi Gao
- Department of Orthopaedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Pengzhen Cheng
- Department of Orthopaedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Bin Liu
- Department of Orthopaedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Sheng Miao
- Department of Orthopaedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Long Bi
- Department of Orthopaedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Liu Yang
- Department of Orthopaedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Guoxian Pei
- Department of Orthopaedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
70
|
Splenic artery denervation: target micro-anatomy, feasibility, and early preclinical experience. Transl Res 2019; 213:100-111. [PMID: 31415732 DOI: 10.1016/j.trsl.2019.07.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 07/09/2019] [Accepted: 07/22/2019] [Indexed: 01/28/2023]
Abstract
This study sought to evaluate perisplenic artery nerve distribution and the feasibility of splenic artery denervation (SDN). The NEXION radiofrequency catheter was used to perform SDN in healthy and inflammatory arthritis pigs. Splenic artery anatomy, nerve distribution, and splenic norepinephrine (NEPI) levels were evaluated before and after SDN. Perisplenic artery nerves were primarily distributed within 2.5 mm of the arterial lumen and were largely sympathetic on the basis of tyrosine hydroxylase expression. The pancreas, tended to be circumferentially positioned around the proximal splenic artery, typically >2.5 mm from the lumen, ensuring that most of the nerves could be targeted without affecting this sensitive organ. The mid segment of the splenic artery was relatively free of contact with the adjacent pancreas. Splenic NEPI levels and nerve abundance followed a decreasing gradient from the proximal to distal splenic artery. SDN resulted in significant reductions in splenic NEPI levels at day 14 (60.7%, P = 0.024) in naïve pigs and day 45 (100%, P = 0.001) in inflammatory arthritis pigs. There was no significant effect of SDN on joint soft tissue injury or circulating inflammatory markers in the inflammatory arthritis model. The majority of perisplenic arterial nerves are within close proximity of the lumen and are primarily sympathetic efferent fibers. Nerves in the mid-segment may be the preferred SDN target given their proximity to the artery and paucity of periarterial off-target organs. SDN appears safe and effective at reducing splenic NEPI levels.
Collapse
|
71
|
Tanaka S, Hammond B, Rosin DL, Okusa MD. Neuroimmunomodulation of tissue injury and disease: an expanding view of the inflammatory reflex pathway. Bioelectron Med 2019; 5:13. [PMID: 32232102 PMCID: PMC7098254 DOI: 10.1186/s42234-019-0029-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 07/09/2019] [Indexed: 02/07/2023] Open
Abstract
Neuroimmunomodulation through peripheral nerve activation is an important therapeutic approach to various disorders. Central to this approach is the inflammatory reflex pathway in which the cholinergic anti-inflammatory pathway represents the efferent limb. Recent studies provide a framework for understanding this control pathway, however our understanding remains incomplete. Genetically modified mice, using optogenetics and pharmacogenomics, have been invaluable resources that will allow investigators to disentangle neural pathways that provide a unifying mechanism by which vagal nerve stimulation (and other means of stimulating the pathway) leads to an anti-inflammatory and tissue protective effect. In this review we describe disease models that contribute to our understanding of how vagal nerve stimulation attenuates inflammation and organ injury: acute kidney injury, rheumatoid arthritis, and inflammatory gastrointestinal disease. The gut microbiota contributes to health and disease and the potential role of the vagus nerve in affecting the relationship between gut microbiota and the immune system and modifying diseases remains an intriguing opportunity to attenuate local and systemic inflammation that undergird disease processes.
Collapse
Affiliation(s)
- Shinji Tanaka
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, University of Virginia, Charlottesville, Virginia USA
| | | | - Diane L. Rosin
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia USA
| | - Mark D. Okusa
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, University of Virginia, Charlottesville, Virginia USA
| |
Collapse
|
72
|
Krishnan S, Lawrence CB. Old Dog New Tricks; Revisiting How Stroke Modulates the Systemic Immune Landscape. Front Neurol 2019; 10:718. [PMID: 31312180 PMCID: PMC6614437 DOI: 10.3389/fneur.2019.00718] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 06/18/2019] [Indexed: 12/27/2022] Open
Abstract
Infections in the post-acute phase of cerebral ischaemia impede optimal recovery by exacerbating morbidity and mortality. Our review aims to reconcile the increased infection susceptibility of patients post-stroke by consolidating our understanding of compartmentalised alterations to systemic immunity. Mounting evidence has catalogued alterations to numerous immune cell populations but an understanding of the mechanisms of long-range communication between the immune system, nervous system and other organs beyond the involvement of autonomic signalling is lacking. By taking our cues from established and emerging concepts of neuro-immune interactions, immune-mediated inter-organ cross-talk, innate immune training and the role of microbiota-derived signals in central nervous system (CNS) function we will explore mechanisms of how cerebral ischaemia could shape systemic immune function. In this context, we will also discuss a key question: how are immune requirements critical for mediating repair of the ischaemic insult balanced by the need for anti-microbial immunity post-stroke, given that they are mediated by mutually exclusive immune networks? Our reformed understanding of the immune landscape post-stroke and novel mechanisms at play could guide targeted therapeutic interventions and initiate a step-change in the clinical management of these infectious complications post-stroke.
Collapse
Affiliation(s)
- Siddharth Krishnan
- Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom.,Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, United Kingdom.,Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Catherine B Lawrence
- Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom.,Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
73
|
Konkel JE, O'Boyle C, Krishnan S. Distal Consequences of Oral Inflammation. Front Immunol 2019; 10:1403. [PMID: 31293577 PMCID: PMC6603141 DOI: 10.3389/fimmu.2019.01403] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 06/03/2019] [Indexed: 12/11/2022] Open
Abstract
Periodontitis is an incredibly prevalent chronic inflammatory disease, which results in the destruction of tooth supporting structures. However, in addition to causing tooth and alveolar bone loss, this oral inflammatory disease has been shown to contribute to disease states and inflammatory pathology at sites distant from the oral cavity. Epidemiological and experimental studies have linked periodontitis to the development and/or exacerbation of a plethora of other chronic diseases ranging from rheumatoid arthritis to Alzheimer's disease. Such studies highlight how the inflammatory status of the oral cavity can have a profound impact on systemic health. In this review we discuss the disease states impacted by periodontitis and explore potential mechanisms whereby oral inflammation could promote loss of homeostasis at distant sites.
Collapse
Affiliation(s)
- Joanne E. Konkel
- Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
- Manchester Collaborative Centre for Inflammation Research (MCCIR), University of Manchester, Manchester, United Kingdom
| | - Conor O'Boyle
- Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Siddharth Krishnan
- Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
- Manchester Collaborative Centre for Inflammation Research (MCCIR), University of Manchester, Manchester, United Kingdom
| |
Collapse
|
74
|
Flynn MC, Pernes G, Lee MKS, Nagareddy PR, Murphy AJ. Monocytes, Macrophages, and Metabolic Disease in Atherosclerosis. Front Pharmacol 2019; 10:666. [PMID: 31249530 PMCID: PMC6584106 DOI: 10.3389/fphar.2019.00666] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 05/23/2019] [Indexed: 12/20/2022] Open
Abstract
Atherosclerotic cardiovascular disease (CVD) is a lipid-driven chronic inflammatory disease, in which macrophages are responsible for taking up these lipids and driving disease progression. Over the years, we and others have uncovered key pathways that regulate macrophage number/function and identified how metabolic disorders such as diabetes and obesity, which are common risk factors for CVD, exacerbate these pathways. This ultimately accelerates the progression of atherosclerosis and hinders atherosclerotic regression. In this review, we discuss the different types of macrophages, from monocyte-derived macrophages, local macrophage proliferation, to macrophage-like vascular smooth muscle cells, that contribute to atherosclerosis as well as myeloid-derived suppressor cells that may have anti-atherogenic effects. We will also discuss how diabetes and obesity influence plaque macrophage accumulation and monocyte production (myelopoiesis) to promote atherogenesis as well as an exciting therapeutic target, S100A8/A9, which mediates myelopoiesis in response to both diabetes and obesity, shown to be effective in reducing atherosclerosis in pre-clinical models of diabetes.
Collapse
Affiliation(s)
- Michelle C Flynn
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Gerard Pernes
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Man Kit Sam Lee
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Prabhakara R Nagareddy
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Andrew J Murphy
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| |
Collapse
|
75
|
Cossío I, Lucas D, Hidalgo A. Neutrophils as regulators of the hematopoietic niche. Blood 2019; 133:2140-2148. [PMID: 30898859 PMCID: PMC6524561 DOI: 10.1182/blood-2018-10-844571] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 12/03/2018] [Indexed: 12/22/2022] Open
Abstract
The niche that supports hematopoietic stem and progenitor cells (HSPCs) in the bone marrow is a highly dynamic structure. It maintains core properties of HSPCs in the steady state, and modulates their proliferation and differentiation in response to changing physiological demands or pathological insults. The dynamic and environment-sensing properties of the niche are shared by the innate immune system. Thus, it is not surprising that innate immune cells, including macrophages and neutrophils, are now recognized as important regulators of the hematopoietic niche and, ultimately, of the stem cells from which they derive. This review synthesizes emerging concepts on niche regulation by immune cells, with a particular emphasis on neutrophils. We argue that the unique developmental, circadian, and migratory properties of neutrophils underlie their critical contributions as regulators of the hematopoietic niche.
Collapse
Affiliation(s)
- Itziar Cossío
- Area of Cell and Developmental Biology, Fundación Centro Nacional de Investigaciones Cardiovasculares (CNIC) Carlos III, Madrid, Spain
| | - Daniel Lucas
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; and
| | - Andrés Hidalgo
- Area of Cell and Developmental Biology, Fundación Centro Nacional de Investigaciones Cardiovasculares (CNIC) Carlos III, Madrid, Spain
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillians-Universität, Munich, Germany
| |
Collapse
|
76
|
Abstract
The intricate interplay between the immune and the nervous systems has been steadily unveiled at both cellular and molecular levels. In this issue of Immunity, Vasamsetti et al. (2018) show that sympathetic nerves drive catecholamine signaling from leukocytes, thereby promoting splenic granulocyte macrophage progenitor (GMP) proliferation and differentiation.
Collapse
Affiliation(s)
- Zhongsheng Hu
- Center for Life Sciences, Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China; Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing 100084, China
| | - Wenwen Zeng
- Center for Life Sciences, Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China; Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing 100084, China.
| |
Collapse
|
77
|
Adrenergic Modulation of Hematopoiesis. J Neuroimmune Pharmacol 2019; 15:82-92. [PMID: 30762159 DOI: 10.1007/s11481-019-09840-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 01/31/2019] [Indexed: 01/11/2023]
Abstract
Hematopoiesis produce every day billions of blood cells and takes place in the bone marrow (BM) by the proliferation and differentiation of hematopoietic stem cells (HSC). HSC are found mainly adjacent to the BM vascular sinusoids where endothelial cells and mesenchimal stromal cells promote HSC maintenance by producing a variety of factors. Other cell types that regulate HSC niches include sympathetic nerves, non-myelinating Schwann cells and a variety of mature hematopoietic cells such as macrophages, neutrophils, and megakaryocytes. This review will focus on the role of adrenergic signals, i.e. of catecholamines, in the regulation of the HSC niche. The available evidence is rather controversial possibly due to the fact that adrenergic receptors are expressed by many cellular components of the niche and also by the often neglected observation that catecholamines may be produced and released also by the BM cells themselves. In addition one has to consider that, physiologically, the sympathetic nervous system (SNS) activity follows a circadian rhythmicity as driven by the suprachiasmatic nucleus (SCN) of the hypothalamus but may be also activated by cognitive and non-cognitive environmental stimuli. The adrenergic modulation of hematopoiesis holds a considerable potential for pharmacological therapeutic approaches in a variety of hematopoietic disorders and for HSC transplantation however the complexity of the system demands further studies. Graphical Abstract Sympathetic nerve termini may release NE while mature BM cells may release norepinephrine (NE) and / or epinephrine (E). Both may bind to β-adrenergic receptor (AR) expressed in nestin+MSC in the hematopoietic stem cell (HSC) niche and regulate the physiological trafficking of HSC by modulating the expression of CXCL12 and SCF. Both NE and E may also activate Lin - c-Kit+ Sca-1+ (LKS) cell via another AR. In addition, NE may also signal to α1-AR expressed in pre-B cells which by TGF-β secretion might regulate proliferation of their lymphoid progenitors in an autocrine manner and/or inhibit myeloid progenitors.
Collapse
|
78
|
Mohammadpour H, Bucsek MJ, Hylander BL, Repasky EA. Depression Stresses the Immune Response and Promotes Prostate Cancer Growth. Clin Cancer Res 2019; 25:2363-2365. [PMID: 30670491 DOI: 10.1158/1078-0432.ccr-18-3980] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 01/10/2019] [Accepted: 01/17/2019] [Indexed: 12/21/2022]
Abstract
Depression induces secretion of neuropeptide Y from prostate cancer cells, which, in turn, recruits myeloid-derived suppressor cells (MDSC) to the tumor; tumor cells and MDSCs secrete IL6, which activates STAT3 within cancer cells. Prostate cancer samples from depressed patients reveal a similar phenotype, suggesting new treatment strategies based upon blockade of β2-adrenergic receptors and/or neuropeptide Y.See related article by Cheng et al., p. 2621.
Collapse
Affiliation(s)
- Hemn Mohammadpour
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York.
| | - Mark J Bucsek
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Bonnie L Hylander
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Elizabeth A Repasky
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| |
Collapse
|
79
|
Cheng Y, Tang XY, Li YX, Zhao DD, Cao QH, Wu HX, Yang HB, Hao K, Yang Y. Depression-Induced Neuropeptide Y Secretion Promotes Prostate Cancer Growth by Recruiting Myeloid Cells. Clin Cancer Res 2018; 25:2621-2632. [PMID: 30504424 DOI: 10.1158/1078-0432.ccr-18-2912] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 11/08/2018] [Accepted: 11/28/2018] [Indexed: 11/16/2022]
Abstract
PURPOSE Psychologic depression has been shown to dysregulate the immune system and promote tumor progression. The aim of this study is to investigate how psychologic depression alters the immune profiles in prostate cancer. EXPERIMENTAL DESIGN We used a murine model of depression in Myc-CaP tumor-bearing immunocompetent FVB mice and Hi-myc mice presenting with spontaneous prostate cancer. Transwell migration and coculture assays were used to evaluate myeloid cell trafficking and cytokine profile changes evoked by Myc-CaP cells that had been treated with norepinephrine (NE), a major elevated neurotransmitter in depression. Chemoattractant, which correlated with immune cell infiltration, was screened by RNA-seq. The chemoattractant and immune cell infiltration were further confirmed using clinical samples of patients with prostate cancer with a high score of psychologic depression. RESULTS Psychologic depression predominantly promoted tumor-associated macrophage (TAM) intratumor infiltrations, which resulted from spleen and circulating monocytic myeloid-derived suppressor cell mobilization. Neuropeptide Y (NPY) released from NE-treated Myc-CaP cells promotes macrophage trafficking and IL6 releasing, which activates STAT3 signaling pathway in prostate cancer cells. Clinical specimens from patients with prostate cancer with higher score of depression revealed higher CD68+ TAM infiltration and stronger NPY and IL6 expression. CONCLUSIONS Depression promotes myeloid cell infiltration and increases IL6 levels by a sympathetic-NPY signal. Sympathetic-NPY inhibition may be a promising strategy for patients with prostate cancer with high score of psychologic depression.See related commentary by Mohammadpour et al., p. 2363.
Collapse
Affiliation(s)
- Yan Cheng
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P.R. China.,Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, P.R. China
| | - Xin-Ying Tang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P.R. China.,Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, P.R. China
| | - Yi-Xuan Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P.R. China.,Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, P.R. China
| | - Dan-Dan Zhao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P.R. China.,Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, P.R. China
| | - Qiu-Hua Cao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P.R. China.,Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, P.R. China
| | - Hong-Xi Wu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P.R. China.,Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, P.R. China
| | - Hong-Bao Yang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P.R. China.,Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, P.R. China
| | - Kun Hao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P.R. China.,Key Lab of Drug Metabolism & Pharmacokinetics, China Pharmaceutical University, Nanjing, P.R. China
| | - Yong Yang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P.R. China. .,Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, P.R. China
| |
Collapse
|