1
|
Zhang S, Sun Z, Chen Z, Bi Y, Wei S, Mao Z, Jin J, Ding Y, Wang W. Endothelial YAP/TEAD1-CXCL17 signaling recruits myeloid-derived suppressor cells against liver ischemia-reperfusion injury. Hepatology 2024:01515467-990000000-00768. [PMID: 38407233 DOI: 10.1097/hep.0000000000000773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 01/30/2024] [Indexed: 02/27/2024]
Abstract
BACKGROUND AND AIMS Liver ischemia-reperfusion injury (IRI) is a common complication of liver transplantation and hepatectomy and causes acute liver dysfunction and even organ failure. Myeloid-derived suppressor cells (MDSCs) accumulate and play immunosuppressive function in cancers and inflammation. However, the role of MDSCs in liver IRI has not been defined. APPROACH AND RESULTS We enrolled recipients receiving OLT and obtained the pre-OLT/post-OLT blood and liver samples. The proportions of MDSCs were significantly elevated after OLT and negatively associated with liver damage. In single-cell RNA-sequencing analysis of liver samples during OLT, 2 cell clusters with MDSC-like phenotypes were identified and showed maturation and infiltration in post-OLT livers. In the mouse model, liver IRI mobilized MDSCs and promoted their infiltration in the damaged liver, and intrahepatic MDSCs were possessed with enhanced immunosuppressive function by upregulation of STAT3 signaling. Under treatment with αGr-1 antibody or adoptive transfer MDSCs to change the proportion of MDSCs in vivo, we found that intrahepatic MDSCs alleviated liver IRI-induced inflammation and damage by inhibiting M1 macrophage polarization. Mechanistically, bulk RNA-sequencing analysis and in vivo experiments verified that C-X-C motif chemokine ligand 17 (CXCL17) was upregulated by YAP/TEAD1 signaling and subsequently recruited MDSCs through binding with GPR35 during liver IRI. Moreover, hepatic endothelial cells were the major cells responsible for CXCL17 expression in injured livers, among which hypoxia-reoxygenation stimulation activated the YAP/TEAD1 complex to promote CXCL17 transcription. CONCLUSIONS Endothelial YAP/TEAD1-CXCL17 signaling recruited MDSCs to attenuate liver IRI, providing evidence of therapeutic potential for managing IRI in liver surgery.
Collapse
Affiliation(s)
- Sitong Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, P.R. China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang, P.R. China
- National Innovation Center for Fundamental Research on Cancer Medicine, Hangzhou, Zhejiang, P.R. China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, P.R. China
- ZJU-Pujian Research & Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic Disease, Hangzhou, Zhejiang, P.R. China
| | - Zhongquan Sun
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, P.R. China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang, P.R. China
- National Innovation Center for Fundamental Research on Cancer Medicine, Hangzhou, Zhejiang, P.R. China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, P.R. China
- ZJU-Pujian Research & Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic Disease, Hangzhou, Zhejiang, P.R. China
| | - Zhenhua Chen
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, P.R. China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang, P.R. China
- National Innovation Center for Fundamental Research on Cancer Medicine, Hangzhou, Zhejiang, P.R. China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, P.R. China
- ZJU-Pujian Research & Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic Disease, Hangzhou, Zhejiang, P.R. China
| | - Yanli Bi
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, P.R. China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang, P.R. China
- National Innovation Center for Fundamental Research on Cancer Medicine, Hangzhou, Zhejiang, P.R. China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, P.R. China
- ZJU-Pujian Research & Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic Disease, Hangzhou, Zhejiang, P.R. China
| | - Shenyu Wei
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, P.R. China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang, P.R. China
- National Innovation Center for Fundamental Research on Cancer Medicine, Hangzhou, Zhejiang, P.R. China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, P.R. China
- ZJU-Pujian Research & Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic Disease, Hangzhou, Zhejiang, P.R. China
| | - Zhengwei Mao
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, P.R. China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| | - Jin Jin
- MOE Laboratory of Biosystem Homeostasis and Protection and Life Sciences Institute, Department of Life Science, Zhejiang University, Hangzhou, Zhejiang, P.R.China
| | - Yuan Ding
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, P.R. China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang, P.R. China
- National Innovation Center for Fundamental Research on Cancer Medicine, Hangzhou, Zhejiang, P.R. China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, P.R. China
- ZJU-Pujian Research & Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic Disease, Hangzhou, Zhejiang, P.R. China
| | - Weilin Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, P.R. China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang, P.R. China
- National Innovation Center for Fundamental Research on Cancer Medicine, Hangzhou, Zhejiang, P.R. China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, P.R. China
- ZJU-Pujian Research & Development Center of Medical Artificial Intelligence for Hepatobiliary and Pancreatic Disease, Hangzhou, Zhejiang, P.R. China
| |
Collapse
|
2
|
Amano H, Eshima K, Ito Y, Nakamura M, Kitasato H, Ogawa F, Hosono K, Iwabuchi K, Uematsu S, Akira S, Narumiya S, Majima M. The microsomal prostaglandin E synthase-1/prostaglandin E2 axis induces recovery from ischaemia via recruitment of regulatory T cells. Cardiovasc Res 2023; 119:1218-1233. [PMID: 35986688 PMCID: PMC10411941 DOI: 10.1093/cvr/cvac137] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 07/24/2022] [Accepted: 07/26/2022] [Indexed: 11/13/2022] Open
Abstract
AIMS Microsomal prostaglandin E synthase-1 (mPGES-1)/prostaglandin E2 (PGE2) induces angiogenesis through the prostaglandin E2 receptor (EP1-4). Among immune cells, regulatory T cells (Tregs), which inhibit immune responses, have been implicated in angiogenesis, and PGE2 is known to modulate the function and differentiation of Tregs. We hypothesized that mPGES-1/PGE2-EP signalling could contribute to recovery from ischaemic conditions by promoting the accumulation of Tregs. METHODS AND RESULTS Wild-type (WT), mPGES-1-deficient (mPges-1-/-), and EP4 receptor-deficient (Ep4-/-) male mice, 6-8 weeks old, were used. Hindlimb ischaemia was induced by femoral artery ligation. Recovery from ischaemia was suppressed in mPges-1-/- mice and compared with WT mice. The number of accumulated forkhead box protein P3 (FoxP3)+ cells in ischaemic muscle tissue was decreased in mPges-1-/- mice compared with that in WT mice. Expression levels of transforming growth factor-β (TGF-β) and stromal cell derived factor-1 (SDF-1) in ischaemic tissue were also suppressed in mPges-1-/- mice. The number of accumulated FoxP3+ cells and blood flow recovery were suppressed when Tregs were depleted by injecting antibody against folate receptor 4 in WT mice but not in mPges-1-/- mice. Recovery from ischaemia was significantly suppressed in Ep4-/- mice compared with that in WT mice. Furthermore, mRNA levels of Foxp3 and Tgf-β were suppressed in Ep4-/- mice. Moreover, the number of accumulated FoxP3+ cells in ischaemic tissue was diminished in Ep4-/- mice compared with that in Ep4+/+ mice. CONCLUSION These findings suggested that mPGES-1/PGE2 induced neovascularization from ischaemia via EP4 by promoting the accumulation of Tregs. Highly selective EP4 agonists could be useful for the treatment of peripheral artery disease.
Collapse
Affiliation(s)
- Hideki Amano
- Department of Pharmacology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Koji Eshima
- Department of Immunology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Yoshiya Ito
- Department of Pharmacology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Masaki Nakamura
- Department of Microbiology, Kitasato University School of Allied Health Science, Kanagawa, Japan
| | - Hidero Kitasato
- Department of Microbiology, Kitasato University School of Allied Health Science, Kanagawa, Japan
| | - Fumihiro Ogawa
- Department of Pharmacology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Kanako Hosono
- Department of Pharmacology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Kazuya Iwabuchi
- Department of Immunology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Satoshi Uematsu
- Department of Immunology and Genomics, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Shizuo Akira
- Laboratory of Host Defense, WPI Immunology Frontier Research Center (IFReC), Osaka University, Osaka, Japan
| | - Shuh Narumiya
- Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masataka Majima
- Department of Pharmacology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
- Department of Medical Therapeutics, Kanagawa Institute of Technology, Atsugi, Kanagawa, Japan
| |
Collapse
|
3
|
Inhibition of protein glycosylation is a novel pro-angiogenic strategy that acts via activation of stress pathways. Nat Commun 2020; 11:6330. [PMID: 33303737 PMCID: PMC7730427 DOI: 10.1038/s41467-020-20108-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 11/11/2020] [Indexed: 01/05/2023] Open
Abstract
Endothelial cell (EC) metabolism is thought to be one of the driving forces for angiogenesis. Here we report the identification of the hexosamine D-mannosamine (ManN) as an EC mitogen and survival factor for bovine and human microvascular EC, with an additivity with VEGF. ManN inhibits glycosylation in ECs and induces significant changes in N-glycan and O-glycan profiles. We further demonstrate that ManN and two N-glycosylation inhibitors stimulate EC proliferation via both JNK activation and the unfolded protein response caused by ER stress. ManN results in enhanced angiogenesis in a mouse skin injury model. ManN also promotes angiogenesis in a mouse hindlimb ischemia model, with accelerated limb blood flow recovery compared to controls. In addition, intraocular injection of ManN induces retinal neovascularization. Therefore, activation of stress pathways following inhibition of protein glycosylation can promote EC proliferation and angiogenesis and may represent a therapeutic strategy for treatment of ischemic disorders. Therapeutic angiogenesis has the potential of inducing and maintaining new blood vessels and thus improving outcomes in patients with ischemic disorders. Mannosamine functions as an endothelial cell mitogen/survival factor through activation of stress pathways and might be useful to protect and regenerate the vascular endothelium in a variety of disorders.
Collapse
|
4
|
Kauppinen A, Kaarniranta K, Salminen A. Potential Role of Myeloid-Derived Suppressor Cells (MDSCs) in Age-Related Macular Degeneration (AMD). Front Immunol 2020; 11:384. [PMID: 32265903 PMCID: PMC7099658 DOI: 10.3389/fimmu.2020.00384] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 02/18/2020] [Indexed: 12/23/2022] Open
Abstract
Myeloid cells, such as granulocytes/neutrophils and macrophages, have responsibilities that include pathogen destruction, waste material degradation, or antigen presentation upon inflammation. During persistent stress, myeloid cells can remain partially differentiated and adopt immunosuppressive functions. Myeloid-derived suppressor cells (MDSCs) are primarily beneficial upon restoring homeostasis after inflammation. Because of their ability to suppress adaptive immunity, MDSCs can also ameliorate autoimmune diseases and semi-allogenic responses, e.g., in pregnancy or transplantation. However, immunosuppression is not always desirable. In certain conditions, such as cancer or chronically inflamed tissue, MDSCs prevent restorative immune responses and thereby aggravate disease progression. Age-related macular degeneration (AMD) is the most common disease in Western countries that severely threatens the central vision of aged people. The pathogenesis of this multifactorial disease is not fully elucidated, but inflammation is known to participate in both dry and wet AMD. In this paper, we provide an overview about the potential role of MDSCs in the pathogenesis of AMD.
Collapse
Affiliation(s)
- Anu Kauppinen
- Faculty of Health Sciences, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Kai Kaarniranta
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland.,Department of Ophthalmology, Kuopio University Hospital, Kuopio, Finland
| | - Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
5
|
Nachmany I, Bogoch Y, Sivan A, Amar O, Bondar E, Zohar N, Yakubovsky O, Fainaru O, Klausner JM, Pencovich N. CD11b +Ly6G + myeloid-derived suppressor cells promote liver regeneration in a murine model of major hepatectomy. FASEB J 2019; 33:5967-5978. [PMID: 30730772 DOI: 10.1096/fj.201801733r] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Liver regeneration depends on sequential activation of pathways and cells involving the remaining organ in recovery of mass. Proliferation of parenchyma is dependent on angiogenesis. Understanding liver regeneration-associated neovascularization may be useful for development of clinical interventions. Myeloid-derived suppressor cells (MDSCs) promote tumor angiogenesis and play a role in developmental processes that necessitate rapid vascularization. We therefore hypothesized that the MDSCs could play a role in liver regeneration. Following partial hepatectomy, MDSCs were enriched within regenerating livers, and their depletion led to increased liver injury and postoperative mortality, reduced liver weights, decreased hepatic vascularization, reduced hepatocyte hypertrophy and proliferation, and aberrant liver function. Gene expression profiling of regenerating liver-derived MDSCs demonstrated a large-scale transcriptional response involving key pathways related to angiogenesis. Functionally, enhanced reactive oxygen species production and angiogenic capacities of regenerating liver-derived MDSCs were confirmed. A comparative analysis revealed that the transcriptional response of MDSCs during liver regeneration resembled that of peripheral blood MDSCs during progression of abdominal tumors, suggesting a common MDSC gene expression profile promoting angiogenesis. In summary, our study shows that MDSCs contribute to early stages of liver regeneration possibly by exerting proangiogenic functions using a unique transcriptional program.-Nachmany, I., Bogoch, Y., Sivan, A., Amar, O., Bondar, E., Zohar, N., Yakubovsky, O., Fainaru, O., Klausner, J. M., Pencovich, N. CD11b+Ly6G+ myeloid-derived suppressor cells promote liver regeneration in a murine model of major hepatectomy.
Collapse
Affiliation(s)
- Ido Nachmany
- The Surgical Division, Department of Surgery B, The Laboratory of Molecular Genetics, Hepato-Pancreato-Biliary (HPB) Cancer Research, Tel-Aviv Sourasky Medical Center-The Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Yoel Bogoch
- The Surgical Division, Department of Surgery B, The Laboratory of Molecular Genetics, Hepato-Pancreato-Biliary (HPB) Cancer Research, Tel-Aviv Sourasky Medical Center-The Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Ayelet Sivan
- The Surgical Division, Department of Surgery B, The Laboratory of Molecular Genetics, Hepato-Pancreato-Biliary (HPB) Cancer Research, Tel-Aviv Sourasky Medical Center-The Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Omer Amar
- The Surgical Division, Department of Surgery B, The Laboratory of Molecular Genetics, Hepato-Pancreato-Biliary (HPB) Cancer Research, Tel-Aviv Sourasky Medical Center-The Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Ekaterina Bondar
- The Surgical Division, Department of Surgery B, The Laboratory of Molecular Genetics, Hepato-Pancreato-Biliary (HPB) Cancer Research, Tel-Aviv Sourasky Medical Center-The Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Nitzan Zohar
- The Surgical Division, Department of Surgery B, The Laboratory of Molecular Genetics, Hepato-Pancreato-Biliary (HPB) Cancer Research, Tel-Aviv Sourasky Medical Center-The Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Oran Yakubovsky
- The Surgical Division, Department of Surgery B, The Laboratory of Molecular Genetics, Hepato-Pancreato-Biliary (HPB) Cancer Research, Tel-Aviv Sourasky Medical Center-The Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Ofer Fainaru
- In Vitro Fertilization (IVF) Unit, Department of Obstetrics and Gynecology, Rambam Medical Center, Haifa, Israel
| | - Joseph M Klausner
- The Surgical Division, Department of Surgery B, The Laboratory of Molecular Genetics, Hepato-Pancreato-Biliary (HPB) Cancer Research, Tel-Aviv Sourasky Medical Center-The Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Niv Pencovich
- The Surgical Division, Department of Surgery B, The Laboratory of Molecular Genetics, Hepato-Pancreato-Biliary (HPB) Cancer Research, Tel-Aviv Sourasky Medical Center-The Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
6
|
Recent studies on micro-/nano-sized biomaterials for cancer immunotherapy. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2016. [DOI: 10.1007/s40005-016-0288-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
7
|
Zlatanova I, Pinto C, Silvestre JS. Immune Modulation of Cardiac Repair and Regeneration: The Art of Mending Broken Hearts. Front Cardiovasc Med 2016; 3:40. [PMID: 27790620 PMCID: PMC5063859 DOI: 10.3389/fcvm.2016.00040] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 09/30/2016] [Indexed: 12/15/2022] Open
Abstract
The accumulation of immune cells is among the earliest responses that manifest in the cardiac tissue after injury. Both innate and adaptive immunity coordinate distinct and mutually non-exclusive events governing cardiac repair, including elimination of the cellular debris, compensatory growth of the remaining cardiac tissue, activation of resident or circulating precursor cells, quantitative and qualitative modifications of the vascular network, and formation of a fibrotic scar. The present review summarizes the mounting evidence suggesting that the inflammatory response also guides the regenerative process following cardiac damage. In particular, recent literature has reinforced the central role of monocytes/macrophages in poising the refreshment of cardiomyocytes in myocardial infarction- or apical resection-induced cardiac insult. Macrophages dictate cardiac myocyte renewal through stimulation of preexisting cardiomyocyte proliferation and/or neovascularization. Nevertheless, substantial efforts are required to identify the nature of these macrophage-derived factors as well as the molecular mechanisms engendered by the distinct subsets of macrophages pertaining in the cardiac tissue. Among the growing inflammatory intermediaries that have been recognized as essential player in heart regeneration, we will focus on the role of interleukin (IL)-6 and IL-13. Finally, it is likely that within the mayhem of the injured cardiac tissue, additional types of inflammatory cells, such as neutrophils, will enter the dance to ignite and refresh the broken heart. However, the protective and detrimental inflammatory pathways have been mainly deciphered in animal models. Future research should be focused on understanding the cellular effectors and molecular signals regulating inflammation in human heart to pave the way for the development of factual therapies targeting the inflammatory compartment in cardiac diseases.
Collapse
Affiliation(s)
- Ivana Zlatanova
- UMRS-970, Paris Centre de Recherche Cardiovasculaire, Institut National de la Santé et de la Recherche Médicale (INSERM), Sorbonne Paris Cité, Université Paris Descartes , Paris , France
| | - Cristina Pinto
- UMRS-970, Paris Centre de Recherche Cardiovasculaire, Institut National de la Santé et de la Recherche Médicale (INSERM), Sorbonne Paris Cité, Université Paris Descartes , Paris , France
| | - Jean-Sébastien Silvestre
- UMRS-970, Paris Centre de Recherche Cardiovasculaire, Institut National de la Santé et de la Recherche Médicale (INSERM), Sorbonne Paris Cité, Université Paris Descartes , Paris , France
| |
Collapse
|
8
|
Zhang MJ, Sansbury BE, Hellmann J, Baker JF, Guo L, Parmer CM, Prenner JC, Conklin DJ, Bhatnagar A, Creager MA, Spite M. Resolvin D2 Enhances Postischemic Revascularization While Resolving Inflammation. Circulation 2016; 134:666-680. [PMID: 27507404 DOI: 10.1161/circulationaha.116.021894] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Accepted: 06/24/2016] [Indexed: 12/31/2022]
Abstract
BACKGROUND Resolvins are lipid mediators generated by leukocytes during the resolution phase of inflammation. They have been shown to regulate the transition from inflammation to tissue repair; however, it is unknown whether resolvins play a role in tissue revascularization following ischemia. METHODS We used a murine model of hind limb ischemia (HLI), coupled with laser Doppler perfusion imaging, microcomputed tomography, and targeted mass spectrometry, to assess the role of resolvins in revascularization and inflammation resolution. RESULTS In mice undergoing HLI, we identified resolvin D2 (RvD2) in bone marrow and skeletal muscle by mass spectrometry (n=4-7 per group). We also identified RvD2 in skeletal muscle biopsies from humans with peripheral artery disease. Monocytes were recruited to skeletal muscle during HLI and isolated monocytes produced RvD2 in a lipoxygenase-dependent manner. Exogenous RvD2 enhanced perfusion recovery in HLI and microcomputed tomography of limb vasculature revealed greater volume, with evidence of tortuous arterioles indicative of arteriogenesis (n=6-8 per group). Unlike other treatment strategies for therapeutic revascularization that exacerbate inflammation, RvD2 did not increase vascular permeability, but reduced neutrophil accumulation and the plasma levels of tumor necrosis factor-α and granulocyte macrophage colony-stimulating factor. In mice treated with RvD2, histopathologic analysis of skeletal muscle of ischemic limbs showed more regenerating myocytes with centrally located nuclei. RvD2 enhanced endothelial cell migration in a Rac-dependent manner, via its receptor, GPR18, and Gpr18-deficient mice had an endogenous defect in perfusion recovery following HLI. Importantly, RvD2 rescued defective revascularization in diabetic mice. CONCLUSIONS RvD2 stimulates arteriogenic revascularization during HLI, suggesting that resolvins may be a novel class of mediators that both resolve inflammation and promote arteriogenesis.
Collapse
Affiliation(s)
- Michael J Zhang
- Institute of Molecular Cardiology, Diabetes and Obesity Center, Division of Cardiovascular Medicine, University of Louisville School of Medicine, Louisville, KY
| | - Brian E Sansbury
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School, Harvard Institutes of Medicine, Boston, MA
| | - Jason Hellmann
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School, Harvard Institutes of Medicine, Boston, MA
| | - James F Baker
- Institute of Molecular Cardiology, Diabetes and Obesity Center, Division of Cardiovascular Medicine, University of Louisville School of Medicine, Louisville, KY
| | - Luping Guo
- Institute of Molecular Cardiology, Diabetes and Obesity Center, Division of Cardiovascular Medicine, University of Louisville School of Medicine, Louisville, KY
| | - Caitlin M Parmer
- Vascular Medicine Section, Cardiovascular Division, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Joshua C Prenner
- Vascular Medicine Section, Cardiovascular Division, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Daniel J Conklin
- Institute of Molecular Cardiology, Diabetes and Obesity Center, Division of Cardiovascular Medicine, University of Louisville School of Medicine, Louisville, KY
| | - Aruni Bhatnagar
- Institute of Molecular Cardiology, Diabetes and Obesity Center, Division of Cardiovascular Medicine, University of Louisville School of Medicine, Louisville, KY
| | - Mark A Creager
- Vascular Medicine Section, Cardiovascular Division, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Matthew Spite
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School, Harvard Institutes of Medicine, Boston, MA
| |
Collapse
|
9
|
Whiteford JR, De Rossi G, Woodfin A. Mutually Supportive Mechanisms of Inflammation and Vascular Remodeling. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 326:201-78. [PMID: 27572130 DOI: 10.1016/bs.ircmb.2016.05.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Chronic inflammation is often accompanied by angiogenesis, the development of new blood vessels from existing ones. This vascular response is a response to chronic hypoxia and/or ischemia, but is also contributory to the progression of disorders including atherosclerosis, arthritis, and tumor growth. Proinflammatory and proangiogenic mediators and signaling pathways form a complex and interrelated network in these conditions, and many factors exert multiple effects. Inflammation drives angiogenesis by direct and indirect mechanisms, promoting endothelial proliferation, migration, and vessel sprouting, but also by mediating extracellular matrix remodeling and release of sequestered growth factors, and recruitment of proangiogenic leukocyte subsets. The role of inflammation in promoting angiogenesis is well documented, but by facilitating greater infiltration of leukocytes and plasma proteins into inflamed tissues, angiogenesis can also propagate chronic inflammation. This review examines the mutually supportive relationship between angiogenesis and inflammation, and considers how these interactions might be exploited to promote resolution of chronic inflammatory or angiogenic disorders.
Collapse
Affiliation(s)
- J R Whiteford
- William Harvey Research Institute, Barts and London School of Medicine and Dentistry, Queen Mary College, University of London, London, United Kingdom
| | - G De Rossi
- William Harvey Research Institute, Barts and London School of Medicine and Dentistry, Queen Mary College, University of London, London, United Kingdom
| | - A Woodfin
- Cardiovascular Division, King's College, University of London, London, United Kingdom.
| |
Collapse
|
10
|
He Y, Wang B, Jia B, Guan J, Zeng H, Pan Z. Effects of Adoptive Transferring Different Sources of Myeloid-Derived Suppressor Cells in Mice Corneal Transplant Survival. Transplantation 2016; 99:2102-8. [PMID: 26270448 DOI: 10.1097/tp.0000000000000749] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Adoptively transferring different sources of myeloid-derived suppressor cells (MDSCs) may assist in mice corneal transplant survival. METHODS Allogeneic full thickness corneal transplantation (donor C57BL/6 to recipient Balb/c mice) was performed. Naive myeloid cells, inflammation-induced MDSCs (iMDSCs), and tumor-induced MDSCs (tMDSCs) were purified from bone marrow of naive, cecal ligation and puncture, or tumor-bearing Balb/c mice, respectively. The inhibitory abilities of myeloid cells toward CD4(+) T cell proliferation were accessed by in vitro carboxyfluorescein diacetate, succinimidyl ester (CFSE) assays. Myeloid cells were adoptively transferred to corneal recipients by retroorbital injection after corneal transplantation. Corneal grafts were examined and photographed for a period of 45 days. The growth of corneal graft neovascularization was quantitatively measured by image editing software. Histopathology was performed to evaluate corneal graft inflammation. RESULTS The iMDSCs and tMDSCs significantly inhibited T cell proliferation in vitro and significantly prolonged corneal allograft survival in vivo. Strikingly, iMDSC transferring significantly reduced neovascularization that was comparable to transferring of tMDSCs, without additional immunosuppression. However, additional adoptive transfer of MDSCs did not further ameliorate corneal survival in these allogeneic corneal transplantation mice. CONCLUSIONS Inflammation-induced MDSC transfer could reduce corneal neovascularization and prolong corneal allograft survival.
Collapse
Affiliation(s)
- Yan He
- 1 Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmic and Visual Science Key Laboratory, Beijing, China. 2 Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Eye institute of The Second Xiangya Hospital of Central South University, Changsha, China. 3 Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China. 4 Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, China
| | | | | | | | | | | |
Collapse
|
11
|
Hammers DW, Rybalko V, Merscham-Banda M, Hsieh PL, Suggs LJ, Farrar RP. Anti-inflammatory macrophages improve skeletal muscle recovery from ischemia-reperfusion. J Appl Physiol (1985) 2015; 118:1067-74. [PMID: 25678696 DOI: 10.1152/japplphysiol.00313.2014] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 02/10/2015] [Indexed: 11/22/2022] Open
Abstract
The presence of macrophages (MPs) is essential for skeletal muscle to properly regenerate following injury. The aim of this study was the evaluation of MP profiles and their importance in skeletal muscle recovering from tourniquet-induced ischemia-reperfusion (I/R). Using flow cytometry, we identified two distinct CD11b(+) MP populations that differ in expression of the surface markers Ly-6C and F4/80. These populations are prominent at 3 and 5 days of reperfusion and molecularly correspond to inflammatory and anti-inflammatory MP phenotypes. Sorted MP populations demonstrated high levels of IGF-I expression, and whole muscle post-I/R IGF-I expression strongly correlates with F4/80 expression. This suggests MPs largely influence postinjury IGF-I upregulation. We additionally demonstrate that direct intramuscular injection of FACS-isolated CD11b(+)Ly-6C(lo)F4/80(hi) MPs improves the functional and histological recovery of I/R-affected muscle. Taken together, these data further support the substantial influence of the innate immune system on muscle regeneration and suggest MP-focused therapeutic approaches may greatly facilitate skeletal muscle recovery from substantial injury.
Collapse
Affiliation(s)
- David W Hammers
- Department of Kinesiology, The University of Texas at Austin, Austin, Texas; and
| | - Viktoriya Rybalko
- Department of Kinesiology, The University of Texas at Austin, Austin, Texas; and
| | | | - Pei-Ling Hsieh
- Department of Kinesiology, The University of Texas at Austin, Austin, Texas; and
| | - Laura J Suggs
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Roger P Farrar
- Department of Kinesiology, The University of Texas at Austin, Austin, Texas; and
| |
Collapse
|
12
|
Tong X, Lv G, Huang J, Min Y, Yang L, Lin PC. Gr-1+CD11b+ myeloid cells efficiently home to site of injury after intravenous administration and enhance diabetic wound healing by neoangiogenesis. J Cell Mol Med 2014; 18:1194-202. [PMID: 24645717 PMCID: PMC4112018 DOI: 10.1111/jcmm.12265] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 01/28/2014] [Indexed: 12/20/2022] Open
Abstract
Vascularization is an important factor that affects diabetic wound healing. There is increasing evidence that myeloid cell lineages play a role in neovascularization. In this study, the efficiency of Gr-1+CD11b+ myeloid cells to home to the site of injury and enhance diabetic wound healing by neoangiogenesis after intravenous administration was investigated. Gr-1+CD11b+ myeloid cells were injected into tail vein after establishment of dorsal window chamber, hindlimb ischaemia and ear-punch injury in diabetic or non-diabetic mice. The Gr-1+CD11b+ myeloid cells efficiently homed to the site of injury after intravenous administration and increased neoangiogenesis. The chemokine receptor type 4 (CXCR4) is robustly expressed by Gr-1+CD11b+ myeloid cells. Inhibition of CXCR4 decreases the homing ability of Gr-1+CD11b+ myeloid cells to the site of injury, which indicates that the CXCR4/SDF-1 axis plays an important role in the homing of Gr-1+CD11b+ myeloid cells to the site of injury. In addition, Gr-1+CD11b+ myeloid cells were found to improve blood flow recovery of ischaemic limb and enhance wound healing in diabetic mice by neoangiogenesis after intravenous administration. Taken together, the results of this study suggest that Gr-1+CD11b+ myeloid cells may serve as a potential cell therapy for diabetic wound healing.
Collapse
Affiliation(s)
- Xiaozhe Tong
- Key Laboratory of Medical Tissue Engineering of Liaoning Province, First Affiliated Hospital of Liaoning Medical University, Jinzhou, Liaoning, China; Department of Traditional Chinese Medicine, First Affiliated Hospital of Liaoning Medical University, Jinzhou, Liaoning, China
| | | | | | | | | | | |
Collapse
|
13
|
Tidball JG, Dorshkind K, Wehling-Henricks M. Shared signaling systems in myeloid cell-mediated muscle regeneration. Development 2014; 141:1184-96. [PMID: 24595286 PMCID: PMC3943178 DOI: 10.1242/dev.098285] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Much of the focus in muscle regeneration has been placed on the identification and delivery of stem cells to promote regenerative capacity. As those efforts have advanced, we have learned that complex features of the microenvironment in which regeneration occurs can determine success or failure. The immune system is an important contributor to that complexity and can determine the extent to which muscle regeneration succeeds. Immune cells of the myeloid lineage play major regulatory roles in tissue regeneration through two general, inductive mechanisms: instructive mechanisms that act directly on muscle cells; and permissive mechanisms that act indirectly to influence regeneration by modulating angiogenesis and fibrosis. In this article, recent discoveries that identify inductive actions of specific populations of myeloid cells on muscle regeneration are presented, with an emphasis on how processes in muscle and myeloid cells are co-regulated.
Collapse
Affiliation(s)
- James G. Tidball
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, CA 90095-1606, USA
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095-1606, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA 90095-1732, USA
| | - Kenneth Dorshkind
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, CA 90095-1606, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA 90095-1732, USA
| | - Michelle Wehling-Henricks
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095-1606, USA
| |
Collapse
|
14
|
Huang J, Lv G, Min Y, Yang L, Lin PC. Intravenous administration of Gr-1+CD11b+ myeloid cells increases neovascularization and improves cardiac function after heart infarction. Int J Cardiol 2013; 168:1702-5. [PMID: 23601210 DOI: 10.1016/j.ijcard.2013.03.069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 03/23/2013] [Indexed: 11/28/2022]
Affiliation(s)
- Jianhua Huang
- Key Laboratory of Medical Tissue Engineering of Liaoning Province, First Affiliated Hospital of Liaoning Medical University, Jinzhou, Liaoning, 121000, China; Department of Radiation Oncology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Surgical Laboratory, First Affiliated Hospital of Liaoning Medical University, Jinzhou, Liaoning, 121000, China.
| | | | | | | | | |
Collapse
|
15
|
Unique expression patterns associated with preferential recruitment of immature myeloid cells into angiogenic versus dormant tumors. Genes Immun 2013; 14:90-8. [PMID: 23303247 DOI: 10.1038/gene.2012.59] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Cancer progression from microscopic dormant tumors into disseminated disease involves tumor angiogenesis, and is commonly referred to as the 'angiogenic switch'. CD11b(+)Gr1(+) immature myeloid cells (IMCs) were reported to promote angiogenesis and tumor progression. Here, we studied a model of tumor dormancy, in which Lewis Lung Carcinoma tumor cells were inoculated intra-abdominally into C57Bl/6J mice. Dormancy versus expansive growth was determined by the site of tumor implantation (lower vs upper abdomen). Global gene expression of IMCs was evaluated in different stages of recruitment, starting in the bone marrow, followed by the peripheral blood and finally in the vascular versus dormant tumors. We first demonstrated a ∼3 fold enrichment of IMCs within vascular tumors as compared with dormant tumors, correlating with tumor-infiltrating CD31(+) endothelial cells. Although their migration from the PB into dormant tumors led to differential expression of a relatively small number of genes, recruitment of IMCs into the upper tumors was associated with a profound transcriptional response. Importantly, a large set of proangiogenic genes were significantly upregulated in IMCs derived from vascular tumors compared with those derived from dormant tumors. We therefore, suggest that proangiogenic versus nonangiogenic transcriptional patterns is associated with the ability of IMCs to promote tumor angiogenesis.
Collapse
|
16
|
Bickert T, Marshall RP, Zhang Z, Ludewig P, Binder M, Klinke A, Rottbauer W, Amling M, Wagener C, Ito WD, Horst AK. Acceleration of Collateral Development by Carcinoembryonic Antigen-Related Cell Adhesion Molecule 1 Expression on CD11b
+
/Gr-1
+
Myeloid Cells—Brief Report. Arterioscler Thromb Vasc Biol 2012; 32:2566-8. [DOI: 10.1161/atvbaha.112.300015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Thomas Bickert
- From the Institute of Clinical Chemistry, Diagnostic Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (T.B., P.L., C.W., A.K.H.); Institute of Osteology and Biomechanics, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (R.P.M., M.A.); Department of Oncology and Hematology, BMT with Section Pneumology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hubertus Wald Tumorzentrum, Hamburg, Germany (M.B.)
| | - Robert Percy Marshall
- From the Institute of Clinical Chemistry, Diagnostic Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (T.B., P.L., C.W., A.K.H.); Institute of Osteology and Biomechanics, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (R.P.M., M.A.); Department of Oncology and Hematology, BMT with Section Pneumology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hubertus Wald Tumorzentrum, Hamburg, Germany (M.B.)
| | - Ziyang Zhang
- From the Institute of Clinical Chemistry, Diagnostic Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (T.B., P.L., C.W., A.K.H.); Institute of Osteology and Biomechanics, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (R.P.M., M.A.); Department of Oncology and Hematology, BMT with Section Pneumology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hubertus Wald Tumorzentrum, Hamburg, Germany (M.B.)
| | - Peter Ludewig
- From the Institute of Clinical Chemistry, Diagnostic Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (T.B., P.L., C.W., A.K.H.); Institute of Osteology and Biomechanics, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (R.P.M., M.A.); Department of Oncology and Hematology, BMT with Section Pneumology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hubertus Wald Tumorzentrum, Hamburg, Germany (M.B.)
| | - Mascha Binder
- From the Institute of Clinical Chemistry, Diagnostic Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (T.B., P.L., C.W., A.K.H.); Institute of Osteology and Biomechanics, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (R.P.M., M.A.); Department of Oncology and Hematology, BMT with Section Pneumology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hubertus Wald Tumorzentrum, Hamburg, Germany (M.B.)
| | - Anna Klinke
- From the Institute of Clinical Chemistry, Diagnostic Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (T.B., P.L., C.W., A.K.H.); Institute of Osteology and Biomechanics, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (R.P.M., M.A.); Department of Oncology and Hematology, BMT with Section Pneumology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hubertus Wald Tumorzentrum, Hamburg, Germany (M.B.)
| | - Wolfgang Rottbauer
- From the Institute of Clinical Chemistry, Diagnostic Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (T.B., P.L., C.W., A.K.H.); Institute of Osteology and Biomechanics, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (R.P.M., M.A.); Department of Oncology and Hematology, BMT with Section Pneumology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hubertus Wald Tumorzentrum, Hamburg, Germany (M.B.)
| | - Michael Amling
- From the Institute of Clinical Chemistry, Diagnostic Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (T.B., P.L., C.W., A.K.H.); Institute of Osteology and Biomechanics, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (R.P.M., M.A.); Department of Oncology and Hematology, BMT with Section Pneumology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hubertus Wald Tumorzentrum, Hamburg, Germany (M.B.)
| | - Christoph Wagener
- From the Institute of Clinical Chemistry, Diagnostic Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (T.B., P.L., C.W., A.K.H.); Institute of Osteology and Biomechanics, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (R.P.M., M.A.); Department of Oncology and Hematology, BMT with Section Pneumology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hubertus Wald Tumorzentrum, Hamburg, Germany (M.B.)
| | - Wulf D. Ito
- From the Institute of Clinical Chemistry, Diagnostic Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (T.B., P.L., C.W., A.K.H.); Institute of Osteology and Biomechanics, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (R.P.M., M.A.); Department of Oncology and Hematology, BMT with Section Pneumology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hubertus Wald Tumorzentrum, Hamburg, Germany (M.B.)
| | - Andrea Kristina Horst
- From the Institute of Clinical Chemistry, Diagnostic Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (T.B., P.L., C.W., A.K.H.); Institute of Osteology and Biomechanics, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (R.P.M., M.A.); Department of Oncology and Hematology, BMT with Section Pneumology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hubertus Wald Tumorzentrum, Hamburg, Germany (M.B.)
| |
Collapse
|
17
|
Abstract
Donepezil {(RS)-2-[(1-benzyl-4-piperidyl)methyl]-5,6-dimethoxy-2,3-dihydroinden-1-one} is a reversible acetylcholinesterase inhibitor and used for treatment of patients with AD (Alzheimer's disease). Recent studies showed that treatment with donepezil reduced production of inflammatory cytokines in PBMCs (peripheral blood mononuclear cells). It was also reported that muscle-derived inflammatory cytokines play a critical role in neovascularization in a hindlimb ischaemia model. We sought to determine whether donepezil affects angiogenesis. A hindlimb ischaemia model was created by unilateral femoral artery ligation. Blood flow recovery examined by laser Doppler perfusion imaging and capillary density by immunohistochemical staining of CD31-positive cells in the ischaemic hindlimb were significantly decreased in donepezil- and physostigmine-treated mice compared with control mice after 2 weeks. Donepezil reduced expression of IL (interleukin)-1β and VEGF (vascular endothelial growth factor) in the ischaemic hindlimb. Intramuscular injections of IL-1β to the ischaemic hindlimb reversed the donepezil-induced VEGF down-regulation and the anti-angiogenic effect. Hypoxia induced IL-1β expression in C2C12 myoblast cells, which was inhibited by pre-incubation with ACh (acetylcholine) or LY294002, a PI3K (phosphoinositide 3-kinase) inhibitor. Donepezil inhibited phosphorylation of Akt [also known as PKB (protein kinase B)], a downstream kinase of PI3K, in the ischaemic hindlimb. These findings suggest that cholinergic stimulation by acetylcholinesterase inhibitors suppresses angiogenesis through inhibition of PI3K-mediated IL-1β induction, which is followed by reduction of VEGF expression. Acetylcholinesterase inhibitor may be a novel anti-angiogenic therapy.
Collapse
|
18
|
Lee DH, Wolstein JM, Pudasaini B, Plotkin M. INK4a deletion results in improved kidney regeneration and decreased capillary rarefaction after ischemia-reperfusion injury. Am J Physiol Renal Physiol 2011; 302:F183-91. [PMID: 21957175 DOI: 10.1152/ajprenal.00407.2011] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The molecular mechanisms that lead to tubular atrophy, capillary loss, and fibrosis following acute kidney injury are not very clear but may involve cell cycle inhibition by increased expression of cyclin kinase inhibitors. The INK4a/ARF locus encodes overlapping genes for two proteins, a cyclin kinase inhibitor, p16(INK4a), and a p53 stabilizer, p19(ARF), from independent promoters. To determine if decreased INK4a gene expression results in improved kidney regeneration, INK4a knockout (KO) and wild-type (WT) mice were subjected to ischemia-reperfusion injury (IRI). p16(INK4a) and p19(ARF) levels were increased markedly in WT mice at 1-28 days after injury. Kidneys were examined to determine the localization and levels of p16(INK4a), apoptosis, cell proliferation, and capillary rarefaction. KO mice displayed decreased tubular cell apoptosis, increased cell proliferation, and lower creatinine levels after injury. KO mice had significantly higher capillary density compared with WT mice at 14-42 days after IRI. Plasma granulocyte colony-stimulating factor (G-CSF) increased after ischemia in both WT and KO mice and was elevated markedly in KO compared with WT mice. KO kidney digests contained higher counts of Gr-1(+)/Cd11b(+) myeloid cells by flow cytometry. KO mice treated with a Gr-1-depleting antibody displayed reduced vascular endothelial growth factor mRNA, plasma G-CSF, and capillary density, and an increase in serum creatinine and medullary myofibroblasts, compared with untreated KO mice 14 days after ischemia. The anti-angiogenic effect of Gr-1 depletion in KO mice was confirmed by Matrigel angiogenesis assays. These results suggest that the absence of p16(INK4a) and p19(ARF) following IRI has a protective effect on the kidney through improved epithelial and microvascular repair, in part by enhancing the mobilization of myeloid cells into the kidney.
Collapse
Affiliation(s)
- David H Lee
- Department of Medicine, New York Medical College, Valhalla, New York, USA
| | | | | | | |
Collapse
|
19
|
Xie J, Liao Y, Yang L, Wu J, Liu C, Xuan W, Li M, Zhang L, Liu Y, Wu P, Bin J. Ultrasound molecular imaging of angiogenesis induced by mutant forms of hypoxia-inducible factor-1α. Cardiovasc Res 2011; 92:256-66. [DOI: 10.1093/cvr/cvr229] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
20
|
Immature myeloid cells accumulate in mouse placenta and promote angiogenesis. Am J Obstet Gynecol 2011; 204:544.e18-23. [PMID: 21420066 DOI: 10.1016/j.ajog.2011.01.060] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2010] [Revised: 01/25/2011] [Accepted: 01/31/2011] [Indexed: 12/17/2022]
Abstract
OBJECTIVE We sought to determine whether CD11b(+)Gr1(+) immature myeloid cells (IMCs), which have been shown to promote tumor angiogenesis, accumulate in the placenta and similarly contribute to blood vessel formation. STUDY DESIGN Experiments were performed on 6- to 8-week-old C57Bl/6J female mice. Placentas from pregnant mice or B16F10 tumors that were subcutaneously implanted were analyzed by flow cytometry and confocal microscopy. To determine the proangiogenic potential of IMCs, Matrigel plug assays were performed. RESULTS IMCs infiltrate the placenta in the proximity of blood vessels, reaching peak concentration at midpregnancy. When isolated from either placentas or B16F10 melanoma tumors, IMCs actively promoted endothelial cell migration into Matrigel plugs in vivo. Furthermore, placental IMCs, similar to tumor-derived IMCs, expressed matrix metalloproteinase-9 and Bv8, 2 pivotal proangiogenic proteins. CONCLUSION IMCs that express matrix metalloproteinase-9 and Bv8 infiltrate placentas of pregnant mice and actively promote angiogenesis. These cells show striking similarity to IMCs that populate malignant tumors.
Collapse
|
21
|
Yurdakul P, Dalton J, Beattie L, Brown N, Erguven S, Maroof A, Kaye PM. Compartment-specific remodeling of splenic micro-architecture during experimental visceral leishmaniasis. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:23-9. [PMID: 21703391 PMCID: PMC3123882 DOI: 10.1016/j.ajpath.2011.03.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Revised: 03/10/2011] [Accepted: 03/21/2011] [Indexed: 12/14/2022]
Abstract
Progressive splenomegaly is a hallmark of visceral leishmaniasis in humans, canids, and rodents. In experimental murine visceral leishmaniasis, splenomegaly is accompanied by pronounced changes in microarchitecture, including expansion of the red pulp vascular system, neovascularization of the white pulp, and remodeling of the stromal cell populations that define the B-cell and T-cell compartments. Here, we show that Ly6C/G+ (Gr-1+) cells, including neutrophils and inflammatory monocytes, accumulate in the splenic red pulp during infection. Cell depletion using monoclonal antibody against either Ly6C/G+ (Gr-1; RB6) or Ly6G+ (1A8) cells increased parasite burden. In contrast, depletion of Ly6C/G+ cells, but not Ly6G+ cells, halted the progressive remodeling of Meca-32+ and CD31+ red pulp vasculature. Strikingly, neither treatment affected white pulp neovascularization or the remodeling of the fibroblastic reticular cell and follicular dendritic cell networks. These findings demonstrate a previously unrecognized compartment-dependent selectivity to the process of splenic vascular remodeling during experimental murine visceral leishmaniasis, attributable to Ly6C+ inflammatory monocytes.
Collapse
Affiliation(s)
- Pinar Yurdakul
- Centre for Immunology and Infection, Hull York Medical School and Department of Biology, University of York, York, United Kingdom
| | | | | | | | | | | | | |
Collapse
|