1
|
Praetzel R, Motaghed M, Fereydouni M, Ahani E, Kepley C. Description and Characterization of Three-Dimensional Human Mast Cell Progenitor Spheroids In Vitro. Cureus 2024; 16:e53708. [PMID: 38455803 PMCID: PMC10919245 DOI: 10.7759/cureus.53708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 01/28/2024] [Indexed: 03/09/2024] Open
Abstract
Human mast cells (MC) are an essential component of the immune system as they uniquely store and release a wide range of soluble mediators through IgE and non-IgE mechanisms. Several tissue sources can be used to differentiate functional MC for in vitro and in vivo studies. Here we describe an improved method for obtaining large numbers of human MC from adipose tissue with advantages over current methods. We analyzed donor parameters (e.g. age, race) on MC-isolation following adipose and skin tissue digestion from healthy donors. Adipose and skin-derived MC were morphologically and immunophenotypically similar in all donors regardless of age. However, donor-dependent variations in MC numbers were observed following tissue digestion. In addition, we identified and characterized three-dimensional structures from which mature MC emerged in vitro using peripheral blood and human tissue sources. MC progenitor spheroids (MCPS) appeared approximately one week following progenitor isolation and were consistently observed to have mature MC attached, emerging, or nearby when cultured in a stem cell factor-containing medium. The overall characteristics of the MCPS were similar from each tissue source. We propose that these MCPS serve as the common source of human MC in vitro.
Collapse
Affiliation(s)
- Rebecca Praetzel
- Department of Molecular and Cellular Sciences, Liberty University College of Osteopathic Medicine, Lynchburg, USA
| | - Mona Motaghed
- Department of Nanoengineering, North Carolina Agricultural and Technical State University, Greensboro, USA
| | - Mohammad Fereydouni
- Department of Nanoscience, University of North Carolina at Greensboro, Greensboro, USA
| | - Elnaz Ahani
- Department of Nanoengineering, North Carolina Agricultural and Technical State University, Greensboro, USA
| | - Chris Kepley
- Department of Molecular and Cellular Sciences, Liberty University College of Osteopathic Medicine, Lynchburg, USA
| |
Collapse
|
2
|
Tsukada A, Takata K, Aikawa J, Iwase D, Mukai M, Uekusa Y, Metoki Y, Inoue G, Miyagi M, Takaso M, Uchida K. Association between High HbA1c Levels and Mast Cell Phenotype in the Infrapatellar Fat Pad of Patients with Knee Osteoarthritis. Int J Mol Sci 2024; 25:877. [PMID: 38255949 PMCID: PMC10815720 DOI: 10.3390/ijms25020877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/02/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Diabetes mellitus (DM) has been suggested as a potential risk factor for knee osteoarthritis (KOA), and its underlying mechanisms remain unclear. The infrapatellar fat pad (IPFP) contributes to OA through inflammatory mediator secretion. Mast cells' (MCs) role in diabetic IPFP pathology is unclear. In 156 KOA patients, hemoglobin A1c (HbA1c) was stratified (HbA1c ≥ 6.5, n = 28; HbA1c < 6.5, n = 128). MC markers (TPSB2, CPA3) in IPFP were studied. Propensity-matched cohorts (n = 27 each) addressed demographic differences. MC-rich fraction (MC-RF) and MC-poor fraction (MC-PF) were isolated, comparing MC markers and genes elevated in diabetic skin-derived MC (PAXIP1, ARG1, HAS1, IL3RA). TPSB2 and CPA3 expression were significantly higher in HbA1c ≥ 6.5 vs. <6.5, both before and after matching. MC-RF showed higher TPSB2 and CPA3 expression than MC-PF in both groups. In the HbA1c ≥ 6.5 group, PAXIP1 and ARG1 expression were significantly higher in the MC-RF than MC-PF. However, no statistical difference in the evaluated genes was detected between the High and Normal groups in the MC-RF. Elevated TPSB2 and CPA3 levels in the IPFP of high HbA1c patients likely reflect higher numbers of MCs in the IPFP, though no difference was found in MC-specific markers on a cell-to-cell basis, as shown in the MC-RF comparison. These findings deepen our understanding of the intricate interplay between diabetes and KOA, guiding targeted therapeutic interventions.
Collapse
Affiliation(s)
- Ayumi Tsukada
- Department of Orthopedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku Kitasato, Sagamihara City 252-0374, Kanagawa, Japan; (A.T.); (K.T.); (J.A.); (D.I.); (M.M.); (Y.U.); (Y.M.); (G.I.); (M.M.); (M.T.)
| | - Ken Takata
- Department of Orthopedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku Kitasato, Sagamihara City 252-0374, Kanagawa, Japan; (A.T.); (K.T.); (J.A.); (D.I.); (M.M.); (Y.U.); (Y.M.); (G.I.); (M.M.); (M.T.)
| | - Jun Aikawa
- Department of Orthopedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku Kitasato, Sagamihara City 252-0374, Kanagawa, Japan; (A.T.); (K.T.); (J.A.); (D.I.); (M.M.); (Y.U.); (Y.M.); (G.I.); (M.M.); (M.T.)
| | - Dai Iwase
- Department of Orthopedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku Kitasato, Sagamihara City 252-0374, Kanagawa, Japan; (A.T.); (K.T.); (J.A.); (D.I.); (M.M.); (Y.U.); (Y.M.); (G.I.); (M.M.); (M.T.)
| | - Manabu Mukai
- Department of Orthopedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku Kitasato, Sagamihara City 252-0374, Kanagawa, Japan; (A.T.); (K.T.); (J.A.); (D.I.); (M.M.); (Y.U.); (Y.M.); (G.I.); (M.M.); (M.T.)
| | - Yui Uekusa
- Department of Orthopedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku Kitasato, Sagamihara City 252-0374, Kanagawa, Japan; (A.T.); (K.T.); (J.A.); (D.I.); (M.M.); (Y.U.); (Y.M.); (G.I.); (M.M.); (M.T.)
| | - Yukie Metoki
- Department of Orthopedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku Kitasato, Sagamihara City 252-0374, Kanagawa, Japan; (A.T.); (K.T.); (J.A.); (D.I.); (M.M.); (Y.U.); (Y.M.); (G.I.); (M.M.); (M.T.)
| | - Gen Inoue
- Department of Orthopedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku Kitasato, Sagamihara City 252-0374, Kanagawa, Japan; (A.T.); (K.T.); (J.A.); (D.I.); (M.M.); (Y.U.); (Y.M.); (G.I.); (M.M.); (M.T.)
| | - Masayuki Miyagi
- Department of Orthopedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku Kitasato, Sagamihara City 252-0374, Kanagawa, Japan; (A.T.); (K.T.); (J.A.); (D.I.); (M.M.); (Y.U.); (Y.M.); (G.I.); (M.M.); (M.T.)
| | - Masashi Takaso
- Department of Orthopedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku Kitasato, Sagamihara City 252-0374, Kanagawa, Japan; (A.T.); (K.T.); (J.A.); (D.I.); (M.M.); (Y.U.); (Y.M.); (G.I.); (M.M.); (M.T.)
| | - Kentaro Uchida
- Department of Orthopedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku Kitasato, Sagamihara City 252-0374, Kanagawa, Japan; (A.T.); (K.T.); (J.A.); (D.I.); (M.M.); (Y.U.); (Y.M.); (G.I.); (M.M.); (M.T.)
- Shonan University Medical Sciences Research Institute, Nishikubo 500, Chigasaki City 253-0083, Kanagawa, Japan
| |
Collapse
|
3
|
Mowafi MM, Elbeialy MAK, Abusinna RG. Effect of magnesium sulfate on oxygenation and lung mechanics in morbidly obese patients undergoing bariatric surgery: a prospective double-blind randomized clinical trial. Korean J Anesthesiol 2023; 76:617-626. [PMID: 36539925 PMCID: PMC10718631 DOI: 10.4097/kja.22446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 12/13/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Respiratory mechanics are often significantly altered in morbidly obese patients and magnesium sulfate (MgSO4) is a promising agent for managing several respiratory disorders. This study aimed to examine the effects of MgSO4 infusions on arterial oxygenation and lung mechanics in patients with morbid obesity undergoing laparoscopic bariatric surgery. METHODS Forty patients with morbid obesity aged 21-60 years scheduled for laparoscopic bariatric surgery under general anesthesia were randomly allocated to either the control (normal saline infusion) or MgSO4 group (30 mg/kg lean body weight [LBW] of 10% MgSO4 in 100 ml normal saline intravenously over 30 min as a loading dose, followed by 10 mg/kg LBW/h for 90 min). The primary outcome was intraoperative arterial oxygenation (ΔPaO2/FiO2). Secondary outcomes included intraoperative static and dynamic compliance, dead space, and hemodynamic parameters. RESULTS At 90 min intraoperatively, the Δ PaO2/FiO2 ratio and the Δ dynamic lung compliance were statistically significantly higher in the MgSO4 group (mean ± SE: 16.1 ± 1.0, 95% CI [14.1, 18.1] and 8.4 ± 0.5 ml/cmH2O, 95% CI [7.4, 9.4]), respectively), and the Δ dead space (%) was statistically significantly lower in the MgSO4 group (mean ± SE: -8.0 ± 0.3%, 95% CI [-8.6, -7.4]) (P < 0.001). No significant differences in static compliance were observed. CONCLUSIONS Although MgSO4 significantly preserved arterial oxygenation and maintained dynamic lung compliance and dead space in patients with morbid obesity, the clinical relevance is minimal. This study failed to adequately reflect the clinical importance of these results.
Collapse
Affiliation(s)
- Marwa M. Mowafi
- Department of Anesthesiology, Intensive Care and Pain Management, Ain-Shams University Faculty of Medicine, Cairo, Egypt
| | - Marwa A. K. Elbeialy
- Department of Anesthesiology, Intensive Care and Pain Management, Ain-Shams University Faculty of Medicine, Cairo, Egypt
| | - Rasha Gamal Abusinna
- Department of Anesthesiology, Intensive Care and Pain Management, Ain-Shams University Faculty of Medicine, Cairo, Egypt
| |
Collapse
|
4
|
Molderings GJ, Afrin LB. A survey of the currently known mast cell mediators with potential relevance for therapy of mast cell-induced symptoms. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:2881-2891. [PMID: 37243761 PMCID: PMC10567897 DOI: 10.1007/s00210-023-02545-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 05/18/2023] [Indexed: 05/29/2023]
Abstract
Mast cells (MCs) occupy a central role in immunological as well as non-immunological processes as reflected in the variety of the mediators by which MCs influence other cells. Published lists of MC mediators have all shown only subsets-usually quite small-of the full repertoire. The full repertoire of MC mediators released by exocytosis is comprehensively compiled here for the first time. The compilation of the data is essentially based on the largely cytokine-focused database COPE®, supplemented with data on the expression of substances in human MCs published in several articles, plus extensive research in the PubMed database. Three hundred and ninety substances could be identified as mediators of human MCs which can be secreted into the extracellular space by activation of the MC. This number might still be an underestimate of the actual number of MC mediators since, in principle, all substances produced by MCs can become mediators because of the possibility of their release by diffusion into the extracellular space, mast cell extracellular traps, and intercellular exchange via nanotubules. When human MCs release mediators in inappropriate manners, this may lead to symptoms in any or all organs/tissues. Thus, such MC activation disorders may clinically present with a myriad of potential combinations of symptoms ranging from trivial to disabling or even life-threatening. The present compilation can be consulted by physicians when trying to gain clarity about MC mediators which may be involved in patients with MC disease symptoms refractory to most therapies.
Collapse
Affiliation(s)
- Gerhard J Molderings
- Institute for Human Genetics, University Hospital of Bonn, Venusberg-Campus 1, D-53127, Bonn, Germany.
| | | |
Collapse
|
5
|
Dileepan KN, Raveendran VV, Sharma R, Abraham H, Barua R, Singh V, Sharma R, Sharma M. Mast cell-mediated immune regulation in health and disease. Front Med (Lausanne) 2023; 10:1213320. [PMID: 37663654 PMCID: PMC10470157 DOI: 10.3389/fmed.2023.1213320] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 07/17/2023] [Indexed: 09/05/2023] Open
Abstract
Mast cells are important components of the immune system, and they perform pro-inflammatory as well as anti-inflammatory roles in the complex process of immune regulation in health and disease. Because of their strategic perivascular localization, sensitivity and adaptability to the microenvironment, and ability to release a variety of preformed and newly synthesized effector molecules, mast cells perform unique functions in almost all organs. Additionally, Mast cells express a wide range of surface and cytoplasmic receptors which enable them to respond to a variety of cytokines, chemicals, and pathogens. The mast cell's role as a cellular interface between external and internal environments as well as between vasculature and tissues is critical for protection and repair. Mast cell interactions with different immune and nonimmune cells through secreted inflammatory mediators may also turn in favor of disease promoting agents. First and forefront, mast cells are well recognized for their multifaceted functions in allergic diseases. Reciprocal communication between mast cells and endothelial cells in the presence of bacterial toxins in chronic/sub-clinical infections induce persistent vascular inflammation. We have shown that mast cell proteases and histamine induce endothelial inflammatory responses that are synergistically amplified by bacterial toxins. Mast cells have been shown to exacerbate vascular changes in normal states as well as in chronic or subclinical infections, particularly among cigarette smokers. Furthermore, a potential role of mast cells in SARS-CoV-2-induced dysfunction of the capillary-alveolar interface adds to the growing understanding of mast cells in viral infections. The interaction between mast cells and microglial cells in the brain further highlights their significance in neuroinflammation. This review highlights the significant role of mast cells as the interface that acts as sensor and early responder through interactions with cells in systemic organs and the nervous system.
Collapse
Affiliation(s)
- Kottarappat N. Dileepan
- Division of Allergy, Clinical Immunology and Rheumatology, Department of Medicine, The University of Kansas Medical Center, Kansas City, KS, United States
| | - Vineesh V. Raveendran
- Division of Allergy, Clinical Immunology and Rheumatology, Department of Medicine, The University of Kansas Medical Center, Kansas City, KS, United States
| | - Rishi Sharma
- Department of Medicine, School of Medicine, University of Missouri, Kansas City, MO, United States
| | - Harita Abraham
- Division of Allergy, Clinical Immunology and Rheumatology, Department of Medicine, The University of Kansas Medical Center, Kansas City, KS, United States
| | - Rajat Barua
- Cardiology Section, Kansas City Veterans Affairs Medical Center, Kansas City, MO, United States
| | - Vikas Singh
- Neurology Section, Kansas City Veterans Affairs Medical Center, Kansas City, MO, United States
| | - Ram Sharma
- Research and Development Service, Kansas City Veterans Affairs Medical Center, Kansas City, MO, United States
| | - Mukut Sharma
- Research and Development Service, Kansas City Veterans Affairs Medical Center, Kansas City, MO, United States
- Midwest Veterans’ Biomedical Research Foundation (MVBRF), Kansas City VA Medical Center, Kansas, MO, United States
| |
Collapse
|
6
|
Zhou L, Xu H. Feasibility of exercise therapy for children with asthma: a meta-analysis. Front Cell Dev Biol 2023; 11:1192929. [PMID: 37492220 PMCID: PMC10364120 DOI: 10.3389/fcell.2023.1192929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 05/24/2023] [Indexed: 07/27/2023] Open
Abstract
Background: Although numerous studies have demonstrated the clear benefits of exercise for people with asthma, controversy remains. This study evaluated the effects of sustained exercise types on lung function and quality of life in patients with asthma. Methods: We searched PubMed, EMBASE, Web of Science, Cochrane Library, China National Knowledge Infrastructure (CNKI), and Wanfang database since January 2000 to August 2022 .included randomized controlled trials (RCTs) of asthmatic children intervened with exercise. The outcomes were lung function and asthma-related quality of life. Fixed-effects model (I2≤50%) or random-effects model (I2>50%) was applied to calculate the pooled effects. Funnel plots were quantified to present publication bias, and a P value <0.05 was statistically significant. Results: Eventually, 15 trials conformed to the selection criteria. The exercise group significantly improved lung function (FEV1 and FVC) in asthmatic children compared with the control group. Forced Expiratory Volume in 1 Second (MD = 2.12, 95%CI = 0.70, 3.53; p = 0.003; I2 = 15%); Forced Vital Capacity (MD = 2.78, 95%CI = 1.26, 4.31; p = 0.0004; I2 = 56%). The immune system markers IL-6 and TNF-α, were significantly reduced in the exercise group. Interleukin-6 (MD = -0.49, 95%CI = -0.81, -0.17; p = 0.003; I2=0%); tumor necrosis factor-α (MD = -0.54, 95%CI = -0.92, -0.15; p = 0.006; I2 = 0%). That quality of life (PAQLQ) was significantly improved in children with asthma in the exercise group. PAQLQ-Total score (MD = 1.06, 95%CI = 0.46, 1.66; p = 0.006; I2 = 94%); PAQLQ-Emotional (MD = 0.91, 95%CI = 0.76, 1.06; p<0.00001; I2 = 90%); PAQLQ-symptoms (MD = 0.87, 95%CI = 0.71, 1.02; p<0.00001; I2 = 95%); PAQLQ-activities (MD = 1.20, 95%CI = 0.58, 1.82; p = 0.00001; I2 = 93%). Meta-analysis showed significant improvements in body composition in the exercise group. BMI (MD = -2.42, 95%CI = -4.40, 0.44; p = 0.02; I2 = 85%). Conclusions: This meta-analysis demonstrated the effectiveness of exercise in improving pulmonary function index (FEV1, FVC), immune system (IL-6, TNF-α, Feno), exercise ability (6MWT), body composition (BMI), and quality of life (PAQLQ) in asthmatic children. Asthmatic children should regularly participate in physical exercise.
Collapse
|
7
|
Chia SL, Kapoor S, Carvalho C, Bajénoff M, Gentek R. Mast cell ontogeny: From fetal development to life-long health and disease. Immunol Rev 2023; 315:31-53. [PMID: 36752151 PMCID: PMC10952628 DOI: 10.1111/imr.13191] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Mast cells (MCs) are evolutionarily ancient innate immune cells with important roles in protective immunity against bacteria, parasites, and venomous animals. They can be found in most organs of the body, where they also contribute to normal tissue functioning, for example by engaging in crosstalk with nerves. Despite this, they are most widely known for their detrimental roles in allergy, anaphylaxis, and atopic disease. Just like macrophages, mast cells were conventionally thought to originate from the bone marrow. However, they are already present in fetal tissues before the onset of bone marrow hematopoiesis, questioning this dogma. In recent years, our view of myeloid cell ontogeny has been revised. We now know that the first mast cells originate from progenitors made in the extra-embryonic yolk sac, and later get supplemented with mast cells produced from subsequent waves of hematopoiesis. In most connective tissues, sizeable populations of fetal-derived mast cells persist into adulthood, where they self-maintain largely independently from the bone marrow. These developmental origins are highly reminiscent of macrophages, which are known to have critical functions in development. Mast cells too may thus support healthy development. Their fetal origins and longevity also make mast cells susceptible to genetic and environmental perturbations, which may render them pathological. Here, we review our current understanding of mast cell biology from a developmental perspective. We first summarize how mast cell populations are established from distinct hematopoietic progenitor waves, and how they are subsequently maintained throughout life. We then discuss what functions mast cells may normally have at early life stages, and how they may be co-opted to cause, worsen, or increase susceptibility to disease.
Collapse
Affiliation(s)
- Shin Li Chia
- Institute for Regeneration and Repair, Centre for Inflammation Research & Centre for Reproductive HealthThe University of EdinburghEdinburghUK
| | - Simran Kapoor
- Institute for Regeneration and Repair, Centre for Inflammation Research & Centre for Reproductive HealthThe University of EdinburghEdinburghUK
| | - Cyril Carvalho
- Institute for Regeneration and Repair, Centre for Inflammation Research & Centre for Reproductive HealthThe University of EdinburghEdinburghUK
| | - Marc Bajénoff
- Centre d'Immunologie de Marseille‐Luminy (CIML)MarseilleFrance
| | - Rebecca Gentek
- Institute for Regeneration and Repair, Centre for Inflammation Research & Centre for Reproductive HealthThe University of EdinburghEdinburghUK
| |
Collapse
|
8
|
WU Y, SONG J, MA A, ZHANG Z, CHEN Z, LI S, QI B, JIA Y. Isolation and extraction of glansreginin A from walnut meal and its effect on the proliferation of 3T3-L1 cells. FOOD SCIENCE AND TECHNOLOGY 2023. [DOI: 10.1590/fst.006023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
Affiliation(s)
- Yongling WU
- Beijing Technology and Business University, China
| | - Jiaxin SONG
- Beijing Technology and Business University, China
| | - Aijin MA
- Beijing Technology and Business University, China
| | - Zijie ZHANG
- Beijing Technology and Business University, China
| | - Zhou CHEN
- Beijing Technology and Business University, China
| | - Siting LI
- Beijing Technology and Business University, China
| | - Bing QI
- Hebei Key Laboratory of Walnut Nutritional Function and Processing Technology, China
| | - Yingmin JIA
- Beijing Technology and Business University, China
| |
Collapse
|
9
|
Shi S, Ye L, Yu X, Jin K, Wu W. Focus on mast cells in the tumor microenvironment: Current knowledge and future directions. Biochim Biophys Acta Rev Cancer 2023; 1878:188845. [PMID: 36476563 DOI: 10.1016/j.bbcan.2022.188845] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 12/01/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022]
Abstract
Mast cells (MCs) are crucial cells participating in both innate and adaptive immune processes that play important roles in protecting human health and in the pathophysiology of various diseases, such as allergies, cardiovascular diseases, and autoimmune diseases. In the context of tumors, MCs are a non-negligible population of immune cells in the tumor microenvironment (TME). In most tumor types, MCs accumulate in both the tumor tissue and the surrounding tissue. MCs interact with multiple components of the TME, affecting TME remodeling and the tumor cell fate. However, controversy persists regarding whether MCs contribute to tumor progression or trigger an anti-tumor immune response. This review focuses on the context of the TME to explore the specific properties and functions of MCs and discusses the crosstalk that occurs between MCs and other components of the TME, which affect tumor angiogenesis and lymphangiogenesis, invasion and metastasis, and tumor immunity through different mechanisms. We also anticipate the potential role of MCs in cancer immunotherapy, which might expand upon the success achieved with existing cancer therapies.
Collapse
Affiliation(s)
- Saimeng Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Longyun Ye
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China.
| | - Kaizhou Jin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China.
| | - Weiding Wu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China.
| |
Collapse
|
10
|
St John AL, Rathore APS, Ginhoux F. New perspectives on the origins and heterogeneity of mast cells. Nat Rev Immunol 2023; 23:55-68. [PMID: 35610312 DOI: 10.1038/s41577-022-00731-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2022] [Indexed: 01/06/2023]
Abstract
Mast cells are immune cells of the haematopoietic lineage that are now thought to have multifaceted functions during homeostasis and in various disease states. Furthermore, while mast cells have been known for a long time to contribute to allergic disease in adults, recent studies, mainly in mice, have highlighted their early origins during fetal development and potential for immune functions, including allergic responses, in early life. Our understanding of the imprinting of mast cells by particular tissues of residence and their potential for regulatory interactions with organ systems such as the peripheral immune, nervous and vascular systems is also rapidly evolving. Here, we discuss the origins of mast cells and their diverse and plastic phenotypes that are influenced by tissue residence. We explore how divergent phenotypes and functions might result from both their hard-wired 'nature' defined by their ontogeny and the 'nurture' they receive within specialized tissue microenvironments.
Collapse
Affiliation(s)
- Ashley L St John
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore.
- Department of Pathology, Duke University Medical Center, Durham, NC, USA.
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- SingHealth Duke-NUS Global Health Institute, Singapore, Singapore.
| | - Abhay P S Rathore
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Florent Ginhoux
- Singapore Immunology Network, A*STAR, Singapore, Singapore.
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore.
| |
Collapse
|
11
|
Identification of Adipose Tissue as a Reservoir of Macrophages after Acute Myocardial Infarction. Int J Mol Sci 2022; 23:ijms231810498. [PMID: 36142416 PMCID: PMC9499676 DOI: 10.3390/ijms231810498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/01/2022] [Accepted: 09/06/2022] [Indexed: 11/16/2022] Open
Abstract
Medullary and extra-medullary hematopoiesis has been shown to govern inflammatory cell infiltration and subsequently cardiac remodeling and function after acute myocardial infarction (MI). Emerging evidence positions adipose tissue (AT) as an alternative source of immune cell production. We, therefore, hypothesized that AT could act as a reservoir of inflammatory cells that participate in cardiac homeostasis after MI. To reveal the distinct role of inflammatory cells derived from AT or bone marrow (BM), chimeric mice were generated using standard repopulation assays. We showed that AMI increased the number of AT-derived macrophages in the cardiac tissue. These macrophages exhibit pro-inflammatory characteristics and their specific depletion improved cardiac function as well as decreased infarct size and interstitial fibrosis. We then reasoned that the alteration of AT-immune compartment in type 2 diabetes could, thus, contribute to defects in cardiac remodeling. However, in these conditions, myeloid cells recruited in the infarcted heart mainly originate from the BM, and AT was no longer used as a myeloid cell reservoir. Altogether, we showed here that a subpopulation of cardiac inflammatory macrophages emerges from myeloid cells of AT origin and plays a detrimental role in cardiac remodeling and function after MI. Diabetes abrogates the ability of AT-derived myeloid cells to populate the infarcted heart.
Collapse
|
12
|
Amin M, Fatema K, Arefin S, Hussain F, Bhowmik D, Hossain M. Obesity, a major risk factor for immunity and severe outcomes of COVID-19. Biosci Rep 2021; 41:BSR20210979. [PMID: 34350941 PMCID: PMC8380923 DOI: 10.1042/bsr20210979] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/29/2021] [Accepted: 08/04/2021] [Indexed: 12/15/2022] Open
Abstract
An influenza-like virus named severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is responsible for COVID-19 disease and spread worldwide within a short time. COVID-19 has now become a significant concern for public health. Obesity is highly prevalent worldwide and is considered a risk factor for impairing the adaptive immune system. Although diabetes, hypertension, cardiovascular disease (CVD), and renal failure are considered the risk factors for COVID-19, obesity is not yet well-considered. The present study approaches establishing a systemic association between the prevalence of obesity and its impact on immunity concerning the severe outcomes of COVID-19 utilizing existing knowledge. Overall study outcomes documented the worldwide prevalence of obesity, its effects on immunity, and a possible underlying mechanism covering obesity-related risk pathways for the severe outcomes of COVID-19. Overall understanding from the present study is that being an immune system impairing factor, the role of obesity in the severe outcomes of COVID-19 is worthy.
Collapse
Affiliation(s)
- Mohammad Tohidul Amin
- Department of Pharmacy, Noakhali Science and Technology University, Noakhali-3814, Bangladesh
| | - Kaniz Fatema
- Department of Applied Chemistry and Chemical Engineering, Noakhali Science and Technology University, Noakhlai-3814, Bangladesh
| | - Sayema Arefin
- Department of Pharmacy, Mawlana Bhashani Science and Technology University, Santosh, Tangail-1902, Bangladesh
| | - Fahad Hussain
- Department of Pharmacy, Noakhali Science and Technology University, Noakhali-3814, Bangladesh
| | - Dipty Rani Bhowmik
- Department of Pharmacy, Noakhali Science and Technology University, Noakhali-3814, Bangladesh
| | - Mohammad Salim Hossain
- Department of Pharmacy, Noakhali Science and Technology University, Noakhali-3814, Bangladesh
| |
Collapse
|
13
|
Chen T, Zhang Y, Dong Y, Zhang D, Xia L, Sun X, Li H, Han C, Wang H, Xu G. Mast cell and heparin promote adipogenesis in superficial fascia of rats. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:159024. [PMID: 34389520 DOI: 10.1016/j.bbalip.2021.159024] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 07/13/2021] [Accepted: 08/03/2021] [Indexed: 10/20/2022]
Abstract
Fascial adipocytes are recently identified as a unique population of adipose cells, which have different developmental origins, anatomical locations, cytological and functional characteristics compared with subcutaneous or visceral adipocytes. Superficial fascia in rats (also in pigs but not obviously in mice) contains numbers of lineage committed preadipocytes which possess adipogenic potential in vivo. The present study aimed to investigate the physiological factors that contribute to fascial adipogenesis in rats. We detected that mast cells, adipose progenitor cells, and mature adipocytes distributed in certain fascia areas were closely associated with each other, and numerous heparin-loaded granules released from mast cells were distributed around fascial preadipocytes. The culture supernatants of rat peritoneal mast cells and RBL-2H3 mast cells contained 20-30 μg/ml of heparin, effectively activated PPAR-responsive luciferase activity, promoted mRNA and protein expressions of key adipogenic genes, and hence increased adipogenic differentiation of fascia- or epididymal adipose-derived stromal cells. Adipogenic effects of mast cell supernatants were mimicked by heparin but not by histamine or 5-hydroxytryptamine, and were antagonized by protamine sulfate. Adipogenic effects of heparin may relate to its chain length of glucosamine units, because heparin stimulated stronger adipogenesis than dalteparin and enoxaparin with relatively short chains. In rats, local administration of heparin-loaded microspheres for 30 days induced adipogenesis in local areas of superficial fascia. Our findings suggested that mast cell and its granule heparin could serve as the endogenous physiological factors to initiate and accelerate local adipogenesis in superficial fascia, or in adipose tissue with the fascia naturally embedded inside.
Collapse
Affiliation(s)
- Tongsheng Chen
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, and Peking University Center for Obesity and Metabolic Disease Research, Beijing 100191, China
| | - Yanfei Zhang
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, and Peking University Center for Obesity and Metabolic Disease Research, Beijing 100191, China
| | - Yingyue Dong
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, and Peking University Center for Obesity and Metabolic Disease Research, Beijing 100191, China
| | - Dandan Zhang
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, and Peking University Center for Obesity and Metabolic Disease Research, Beijing 100191, China
| | - Lisha Xia
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, and Peking University Center for Obesity and Metabolic Disease Research, Beijing 100191, China
| | - Xiaozhe Sun
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, and Peking University Center for Obesity and Metabolic Disease Research, Beijing 100191, China
| | - Hanxiao Li
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, and Peking University Center for Obesity and Metabolic Disease Research, Beijing 100191, China
| | - Chunmiao Han
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, and Peking University Center for Obesity and Metabolic Disease Research, Beijing 100191, China
| | - Huamin Wang
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, and Peking University Center for Obesity and Metabolic Disease Research, Beijing 100191, China
| | - Guoheng Xu
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, and Peking University Center for Obesity and Metabolic Disease Research, Beijing 100191, China.
| |
Collapse
|
14
|
Driving regeneration, instead of healing, in adult mammals: the decisive role of resident macrophages through efferocytosis. NPJ Regen Med 2021; 6:41. [PMID: 34344890 PMCID: PMC8333253 DOI: 10.1038/s41536-021-00151-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 07/07/2021] [Indexed: 12/19/2022] Open
Abstract
Tissue repair after lesion usually leads to scar healing and thus loss of function in adult mammals. In contrast, other adult vertebrates such as amphibians have the ability to regenerate and restore tissue homeostasis after lesion. Understanding the control of the repair outcome is thus a concerning challenge for regenerative medicine. We recently developed a model of induced tissue regeneration in adult mice allowing the comparison of the early steps of regenerative and scar healing processes. By using studies of gain and loss of function, specific cell depletion approaches, and hematopoietic chimeras we demonstrate here that tissue regeneration in adult mammals depends on an early and transient peak of granulocyte producing reactive oxygen species and an efficient efferocytosis specifically by tissue-resident macrophages. These findings highlight key and early cellular pathways able to drive tissue repair towards regeneration in adult mammals.
Collapse
|
15
|
Lopez-Perez D, Redruello-Romero A, Garcia-Rubio J, Arana C, Garcia-Escudero LA, Tamayo F, Puentes-Pardo JD, Moreno-SanJuan S, Salmeron J, Blanco A, Galvez J, Leon J, Carazo Á. In Patients With Obesity, the Number of Adipose Tissue Mast Cells Is Significantly Lower in Subjects With Type 2 Diabetes. Front Immunol 2021; 12:664576. [PMID: 34093556 PMCID: PMC8177010 DOI: 10.3389/fimmu.2021.664576] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 05/05/2021] [Indexed: 12/15/2022] Open
Abstract
Type 2 diabetes (T2D) is a rising global health problem mainly caused by obesity and a sedentary lifestyle. In healthy individuals, white adipose tissue (WAT) has a relevant homeostatic role in glucose metabolism, energy storage, and endocrine signaling. Mast cells contribute to these functions promoting WAT angiogenesis and adipogenesis. In patients with T2D, inflammation dramatically impacts WAT functioning, which results in the recruitment of several leukocytes, including monocytes, that enhance this inflammation. Accordingly, the macrophages population rises as the WAT inflammation increases during the T2D status worsening. Since mast cell progenitors cannot arrive at WAT, the amount of WAT mast cells depends on how the new microenvironment affects progenitor and differentiated mast cells. Here, we employed a flow cytometry-based approach to analyze the number of mast cells from omental white adipose tissue (o-WAT) and subcutaneous white adipose tissue (s-WAT) in a cohort of 100 patients with obesity. Additionally, we measured the number of mast cell progenitors in a subcohort of 15 patients. The cohort was divided in three groups: non-T2D, pre-T2D, and T2D. Importantly, patients with T2D have a mild condition (HbA1c <7%). The number of mast cells and mast cell progenitors was lower in patients with T2D in both o-WAT and s-WAT in comparison to subjects from the pre-T2D and non-T2D groups. In the case of mast cells in o-WAT, there were statistically significant differences between non-T2D and T2D groups (p = 0.0031), together with pre-T2D and T2D groups (p=0.0097). However, in s-WAT, the differences are only between non-T2D and T2D groups (p=0.047). These differences have been obtained with patients with a mild T2D condition. Therefore, little changes in T2D status have a huge impact on the number of mast cells in WAT, especially in o-WAT. Due to the importance of mast cells in WAT physiology, their decrease can reduce the capacity of WAT, especially o-WAT, to store lipids and cause hypoxic cell deaths that will trigger inflammation.
Collapse
Affiliation(s)
- David Lopez-Perez
- Department of Pharmacology, Faculty of Pharmacy, University of Granada, Granada, Spain.,Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Anaïs Redruello-Romero
- Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | | | - Carlos Arana
- Endocrinology and Nutrition Unit, Virgen de las Nieves University Hospital, Granada, Spain
| | - Luis A Garcia-Escudero
- Department of Statistics and Operative Research, Faculty of Sciences, University of Valladolid, Valladolid, Spain
| | | | - Jose D Puentes-Pardo
- Department of Pharmacology, Faculty of Pharmacy, University of Granada, Granada, Spain.,Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Sara Moreno-SanJuan
- Cytometry and Microscopy Research Service, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Javier Salmeron
- Gastroenterology Unit, San Cecilio University Hospital, Granada, Spain
| | - Armando Blanco
- Department of Computer Science and Artificial Intelligence, University of Granada, Granada, Spain
| | - Julio Galvez
- Department of Pharmacology, Faculty of Pharmacy, University of Granada, Granada, Spain.,Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain.,Centro de Investigación Biomédica En Red para Enfermedades Hepáticas y Digestivas (CIBER-EHD), Center for Biomedical Research, University of Granada, Granada, Spain
| | - Josefa Leon
- Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Ángel Carazo
- Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain.,Clinical Management Unit of Digestive Disease, San Cecilio University Hospital, Granada, Spain
| |
Collapse
|
16
|
Michailidou Z, Gomez-Salazar M, Alexaki VI. Innate Immune Cells in the Adipose Tissue in Health and Metabolic Disease. J Innate Immun 2021; 14:4-30. [PMID: 33849008 DOI: 10.1159/000515117] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 02/09/2021] [Indexed: 11/19/2022] Open
Abstract
Metabolic disorders, such as obesity, type 2 diabetes mellitus, and nonalcoholic fatty liver disease, are characterized by chronic low-grade tissue and systemic inflammation. During obesity, the adipose tissue undergoes immunometabolic and functional transformation. Adipose tissue inflammation is driven by innate and adaptive immune cells and instigates insulin resistance. Here, we discuss the role of innate immune cells, that is, macrophages, neutrophils, eosinophils, natural killer cells, innate lymphoid type 2 cells, dendritic cells, and mast cells, in the adipose tissue in the healthy (lean) and diseased (obese) state and describe how their function is shaped by the obesogenic microenvironment, and humoral, paracrine, and cellular interactions. Moreover, we particularly outline the role of hypoxia as a central regulator in adipose tissue inflammation. Finally, we discuss the long-lasting effects of adipose tissue inflammation and its potential reversibility through drugs, caloric restriction, or exercise training.
Collapse
Affiliation(s)
- Zoi Michailidou
- Centre for Cardiovascular Sciences, Edinburgh University, Edinburgh, United Kingdom
| | - Mario Gomez-Salazar
- Centre for Cardiovascular Sciences, Edinburgh University, Edinburgh, United Kingdom
| | - Vasileia Ismini Alexaki
- Institute for Clinical Chemistry and Laboratory Medicine, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
17
|
Weinstock LB, Pace LA, Rezaie A, Afrin LB, Molderings GJ. Mast Cell Activation Syndrome: A Primer for the Gastroenterologist. Dig Dis Sci 2021; 66:965-982. [PMID: 32328892 DOI: 10.1007/s10620-020-06264-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 04/08/2020] [Indexed: 02/06/2023]
Abstract
Mast cell activation syndrome is thought to be a common, yet under-recognized, chronic multi-system disorder caused by inappropriate mast cell activation. Gastrointestinal symptoms are frequently reported by these patients and are often mistaken by physicians as functional gastrointestinal disorders. This syndrome can be diagnosed by the medical history and measurable biomarkers. Gastroenterologists manage diseases associated with active inflammatory cells including neutrophils, lymphocytes, macrophages, and eosinophils. The mast cell has only recently been recognized as a major player in our specialty. Gastrointestinal disorders from mast cell mediators often present with apparent irritable bowel syndrome, dyspepsia, chronic or cyclical nausea, and heartburn. Individuals with mast cell activation syndrome experience significant delays in diagnosis. The gastrointestinal symptoms are often refractory to symptom-targeted prescription medications. Beyond avoiding triggers, the best therapy is directed at modulating mast cell activation and the effects of the mediators. Many of these therapies are simple over-the-counter medications. In this article, we review mast cell function and dysfunction and the gastrointestinal symptoms, comorbid conditions, diagnosis, and management of mast cell activation syndrome. Gastroenterologists who become aware of this syndrome can dramatically improve the quality of life for their patients who previously have been labeled with a functional gastrointestinal disorder.
Collapse
Affiliation(s)
- Leonard B Weinstock
- Specialists in Gastroenterology, 11525 Olde Cabin Rd, St. Louis, MO, 63141, USA.
| | - Laura A Pace
- Division of Gastroenterology, Department of Internal Medicine, University of Utah, 30 N 1900 E, SOM 4R118, Salt Lake City, UT, 84132, USA
| | - Ali Rezaie
- Cedars-Sinai Medical Center, Gastroenterology, 8730 Alden Dr., Suite 204E, Los Angeles, CA, 90048, USA
| | - Lawrence B Afrin
- Armonk Integrative Medicine, Hematology/Oncology, 3010 Westchester Avenue, Suite 401, Armonk, NY, 10577, USA
| | | |
Collapse
|
18
|
BOONYARATTANASOONTHORN T, SATO K, OKAMATSU-OGURA Y, MORIMATSU M, AGUI T. The response of adipose tissues to Mycoplasma pulmonis and Sendai virus infection in C57BL/6 and DBA/2 mice. J Vet Med Sci 2021; 83:403-411. [PMID: 33487624 PMCID: PMC8025407 DOI: 10.1292/jvms.20-0625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 01/06/2021] [Indexed: 11/30/2022] Open
Abstract
Adipose tissues in mammals are categorized into white and brown adipose tissues in which cellular morphology, cell functions, and tissue distribution are different. White adipose tissue (WAT) plays a major role in energy reservation, while brown adipose tissue (BAT) mainly relates to the thermoregulation of the body. One interesting function of adipose tissue is the response to the infection, especially the pathogens that cause pneumonia. We have previously reported that DBA/2 (D2) mice are susceptible to pathogens causing pneumonia, Mycoplasma (M.) pulmonis and Sendai virus (SeV), whereas C57BL/6 (B6) mice are resistant to them. Furthermore, morphological alteration of mediastinal fat tissue (MFT) was seen after infection of M. pulmonis in D2 mice but not in B6 mice. In this study, we aimed to exhibit the difference in adipose tissue response in other areas, including interscapular brown adipose tissue (iBAT), inguinal white adipose tissue (ingWAT), and perigonadal WAT (perigoWAT) between resistant strain, B6 and susceptible strain, D2 after challenging them with M. pulmonis and SeV. Compared with B6 mice, D2 mice showed an increase in fat-associated lymphoid cluster in MFT, an increase in BAT in both iBAT and ingWAT after M. pulmonis and SeV infection. The results of this study indicate that pneumonia caused by M. pulmonis and SeV infection induces browning of adipocyte, suggesting that BAT plays a role in pathogen infection and inflammation.
Collapse
Affiliation(s)
- Tussapon BOONYARATTANASOONTHORN
- Laboratory of Laboratory Animal Science and Medicine, Department of Applied Veterinary Sciences, Graduate School of Veterinary
Medicine, Hokkaido University, Hokkaido 060-0818, Japan
- Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Keisuke SATO
- Laboratory of Laboratory Animal Science and Medicine, Department of Applied Veterinary Sciences, Graduate School of Veterinary
Medicine, Hokkaido University, Hokkaido 060-0818, Japan
| | - Yuko OKAMATSU-OGURA
- Laboratory of Biochemistry, Department of Basic Veterinary Sciences, Graduate School of Veterinary Medicine, Hokkaido University,
Hokkaido 060-0818, Japan
| | - Masami MORIMATSU
- Laboratory of Laboratory Animal Science and Medicine, Department of Applied Veterinary Sciences, Graduate School of Veterinary
Medicine, Hokkaido University, Hokkaido 060-0818, Japan
| | - Takashi AGUI
- Laboratory of Laboratory Animal Science and Medicine, Department of Applied Veterinary Sciences, Graduate School of Veterinary
Medicine, Hokkaido University, Hokkaido 060-0818, Japan
| |
Collapse
|
19
|
Gou L, Yue GGL, Puno PT, Lau CBS. A review on the relationship of mast cells and macrophages in breast cancer - Can herbs or natural products facilitate their anti-tumor effects? Pharmacol Res 2020; 164:105321. [PMID: 33285235 DOI: 10.1016/j.phrs.2020.105321] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 11/05/2020] [Accepted: 11/21/2020] [Indexed: 12/27/2022]
Abstract
Breast cancer is an inflammation-related cancer whose tumor microenvironment is largely infiltrated by inflammatory cells. These inflammatory cells including mast cells and macrophages have been elucidated to be vital participants in breast tumor proliferation, survival, invasion and migration. However, the functions of mast cells and macrophages in breast cancer are quite distinct based on recent data. Mast cells exhibit both anti-tumoral and pro-tumoral functions on breast cancer, while high number of tumor-associated macrophages (TAMs) are strongly correlated with poor prognosis and higher risk of distant metastasis in breast cancer patients. Besides, many natural products/extracts have been reported to regulate mast cells and macrophages. In this review, the roles of mast cells and macrophages play in breast cancer are discussed and a summary of those natural products/herbs regulating the functions of mast cells or macrophages is also presented.
Collapse
Affiliation(s)
- Leilei Gou
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Grace Gar-Lee Yue
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, HKSAR, China; State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, New Territories, HKSAR, China
| | - Pema Tenzin Puno
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China.
| | - Clara Bik-San Lau
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, HKSAR, China; State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, New Territories, HKSAR, China.
| |
Collapse
|
20
|
Dong J, Chen L, Zhang Y, Jayaswal N, Mezghani I, Zhang W, Veves A. Mast Cells in Diabetes and Diabetic Wound Healing. Adv Ther 2020; 37:4519-4537. [PMID: 32935286 PMCID: PMC7547971 DOI: 10.1007/s12325-020-01499-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/04/2020] [Indexed: 12/11/2022]
Abstract
Mast cells (MCs) are granulated, immune cells of the myeloid lineage that are present in connective tissues. Apart from their classical role in allergies, MCs also mediate various inflammatory responses due to the nature of their secretory products. They are involved in important physiological and pathophysiological responses related to inflammation, chronic wounds, and autoimmune diseases. There are also indications that MCs are associated with diabetes and its complications. MCs and MC-derived mediators participate in all wound healing stages and are involved in the pathogenesis of non-healing, chronic diabetic foot ulcers (DFUs). More specifically, recent work has shown increased degranulation of skin MCs in human diabetes and diabetic mice, which is associated with impaired wound healing. Furthermore, MC stabilization, either systemic or local at the skin level, improves wound healing in diabetic mice. Understanding the precise role of MCs in wound progression and healing processes can be of critical importance as it can lead to the development of new targeted therapies for diabetic foot ulceration, one of the most devastating complications of diabetes.
Collapse
Affiliation(s)
- Jie Dong
- Joslin-Beth Israel Deaconess Foot Center and The Rongxiang Xu, MD, Center for Regenerative Therapeutics, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Lihong Chen
- Joslin-Beth Israel Deaconess Foot Center and The Rongxiang Xu, MD, Center for Regenerative Therapeutics, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Ying Zhang
- Joslin-Beth Israel Deaconess Foot Center and The Rongxiang Xu, MD, Center for Regenerative Therapeutics, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Navin Jayaswal
- Joslin-Beth Israel Deaconess Foot Center and The Rongxiang Xu, MD, Center for Regenerative Therapeutics, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Ikram Mezghani
- Joslin-Beth Israel Deaconess Foot Center and The Rongxiang Xu, MD, Center for Regenerative Therapeutics, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Weijie Zhang
- Joslin-Beth Israel Deaconess Foot Center and The Rongxiang Xu, MD, Center for Regenerative Therapeutics, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
- LanZhou University of Technology, 287 Langongping Road, Qilihe District, Lanzhou, Gansu, China
| | - Aristidis Veves
- Joslin-Beth Israel Deaconess Foot Center and The Rongxiang Xu, MD, Center for Regenerative Therapeutics, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
21
|
Valent P, Akin C, Hartmann K, Nilsson G, Reiter A, Hermine O, Sotlar K, Sperr WR, Escribano L, George TI, Kluin-Nelemans HC, Ustun C, Triggiani M, Brockow K, Gotlib J, Orfao A, Kovanen PT, Hadzijusufovic E, Sadovnik I, Horny HP, Arock M, Schwartz LB, Austen KF, Metcalfe DD, Galli SJ. Mast cells as a unique hematopoietic lineage and cell system: From Paul Ehrlich's visions to precision medicine concepts. Am J Cancer Res 2020; 10:10743-10768. [PMID: 32929378 PMCID: PMC7482799 DOI: 10.7150/thno.46719] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 08/06/2020] [Indexed: 02/07/2023] Open
Abstract
The origin and functions of mast cells (MCs) have been debated since their description by Paul Ehrlich in 1879. MCs have long been considered 'reactive bystanders' and 'amplifiers' in inflammatory processes, allergic reactions, and host responses to infectious diseases. However, knowledge about the origin, phenotypes and functions of MCs has increased substantially over the past 50 years. MCs are now known to be derived from multipotent hematopoietic progenitors, which, through a process of differentiation and maturation, form a unique hematopoietic lineage residing in multiple organs. In particular, MCs are distinguishable from basophils and other hematopoietic cells by their unique phenotype, origin(s), and spectrum of functions, both in innate and adaptive immune responses and in other settings. The concept of a unique MC lineage is further supported by the development of a distinct group of neoplasms, collectively referred to as mastocytosis, in which MC precursors expand as clonal cells. The clinical consequences of the expansion and/or activation of MCs are best established in mastocytosis and in allergic inflammation. However, MCs have also been implicated as important participants in a number of additional pathologic conditions and physiological processes. In this article, we review concepts regarding MC development, factors controlling MC expansion and activation, and some of the fundamental roles MCs may play in both health and disease. We also discuss new concepts for suppressing MC expansion and/or activation using molecularly-targeted drugs.
Collapse
|
22
|
Elieh Ali Komi D, Shafaghat F, Christian M. Crosstalk Between Mast Cells and Adipocytes in Physiologic and Pathologic Conditions. Clin Rev Allergy Immunol 2020; 58:388-400. [PMID: 32215785 PMCID: PMC7244609 DOI: 10.1007/s12016-020-08785-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Excessive fatty acids and glucose uptake support the infiltration of adipose tissue (AT) by a variety of immune cells including neutrophils, pro-inflammatory M1 macrophages, and mast cells (MCs). These cells promote inflammation by releasing pro-inflammatory mediators. The involvement of MCs in AT biology is supported by their accumulation in the AT of obese individuals along with significantly higher serum levels of MC-derived tryptase. AT-resident MCs under the influence of locally derived adipokines such as leptin become activated and release pro-inflammatory cytokines including TNFα that worsens the inflammatory state. MCs support angiogenesis in AT by releasing chymase and inducing preadipocyte differentiation and also the proliferation of adipocytes through 15-deoxy-delta PGJ2/PPARγ interaction. Additionally, they contribute to the remodeling of the AT extracellular matrix (ECM) and play a role in the recruitment and activation of leukocytes. MC degranulation has been linked to brown adipocyte activation, and evidence indicates an important link between MCs and the appearance of BRITE/beige adipocytes in white AT. Cell crosstalk between MCs and AT-resident cells, mainly adipocytes and immune cells, shows that these cells play a critical role in the regulation of AT homeostasis and inflammation.
Collapse
Affiliation(s)
- Daniel Elieh Ali Komi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farzaneh Shafaghat
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mark Christian
- School of Science and Technology, Nottingham, NG11 8NS, UK.
| |
Collapse
|
23
|
Abstract
Adipose tissue (AT) plays a central role in both metabolic health and pathophysiology. Its expansion in obesity results in increased mortality and morbidity, with contributions to cardiovascular disease, diabetes mellitus, fatty liver disease, and cancer. Obesity prevalence is at an all-time high and is projected to be 50% in the United States by 2030. AT is home to a large variety of immune cells, which are critical to maintain normal tissue functions. For example, γδ T cells are fundamental for AT innervation and thermogenesis, and macrophages are required for recycling of lipids released by adipocytes. The expansion of visceral white AT promotes dysregulation of its immune cell composition and likely promotes low-grade chronic inflammation, which has been proposed to be the underlying cause for the complications of obesity. Interestingly, weight loss after obesity alters the AT immune compartment, which may account for the decreased risk of developing these complications. Recent technological advancements that allow molecular investigation on a single-cell level have led to the discovery of previously unappreciated heterogeneity in many organs and tissues. In this review, we will explore the heterogeneity of immune cells within the visceral white AT and their contributions to homeostasis and pathology.
Collapse
Affiliation(s)
- Ada Weinstock
- Department of Medicine, Leon H. Charney Division of Cardiology, Cardiovascular Research Center, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Hernandez Moura Silva
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, NY, 10016, USA
| | - Kathryn J. Moore
- Department of Medicine, Leon H. Charney Division of Cardiology, Cardiovascular Research Center, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Edward A. Fisher
- Department of Medicine, Leon H. Charney Division of Cardiology, Cardiovascular Research Center, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| |
Collapse
|
24
|
Guo X, Cheng L, Yang S, Che H. Pro-inflammatory immunological effects of adipose tissue and risk of food allergy in obesity: Focus on immunological mechanisms. Allergol Immunopathol (Madr) 2020; 48:306-312. [PMID: 31477390 DOI: 10.1016/j.aller.2019.06.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 05/21/2019] [Accepted: 06/03/2019] [Indexed: 12/12/2022]
Abstract
Over the past three decades, the number of obese people has risen steadily. The chronic low-grade inflammatory state and the non-specific activation of the immune system have contributed greatly to the development of obesity-related immunology. Food allergy as a kind of inflammatory disease with abnormal immune response may be associated with obesity. This review begins with the pro-inflammatory immunological effects of adipose tissue in obesity, and explains the possible effects of obesity on food allergy. In short, obesity not only directly causes imbalance of allergic-related immune cells in adipose tissue, but also indirectly causes this consequence through affecting expression of adipocytokines and peroxisome proliferator-activated receptor gamma (PPARγ) in adipose tissue. As a result, circulating levels of pro-inflammatory factors which are partly derived from adipose tissue increase, which might cause intestinal barrier injury. Therefore, obesity may increase the risk of food allergy.
Collapse
Affiliation(s)
- X Guo
- College of Food Science and Nutritional Engineering, China Agricultural University, China; Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, China
| | - L Cheng
- College of Food Science and Nutritional Engineering, China Agricultural University, China; Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, China
| | - S Yang
- College of Food Science and Nutritional Engineering, China Agricultural University, China; Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, China
| | - H Che
- College of Food Science and Nutritional Engineering, China Agricultural University, China; Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, China.
| |
Collapse
|
25
|
Lin Y, Li X, Fan C, Yang F, Hao D, Ge W, Gao Y. Cardioprotective effects of rat adipose‑derived stem cells differ under normoxic/physioxic conditions and are associated with paracrine factor secretion. Int J Mol Med 2020; 45:1591-1600. [PMID: 32323745 DOI: 10.3892/ijmm.2020.4524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 02/13/2020] [Indexed: 11/06/2022] Open
Abstract
Adipose tissue‑derived stem cells (ASCs) are beneficial for myocardial regeneration. The physiological oxygen content of human organs is estimated to range between 1 and 11%. However, in the majority of previous in vitro studies with cultured ASCs, the O2 concentration was artificially set to 21%. The present study aimed to compare the protective effects of rat ASCs on neonatal rat ventricular myocytes (NRVMs) under normoxic (21% O2) and physioxic (5% O2) conditions. Rat NRVMs cultured under normoxia or physioxia were treated with H2O2 or left untreated, and further co‑cultured with ASCs in 21% or 5% O2. The apoptosis of NRVMs was evaluated by Annexin V staining and quantitating the protein levels of Bcl‑2 and Bax by western blotting. The oxidative stress of NRVMs was determined by a glutathione/oxidized glutathione assay kit. The concentrations of secreted vascular endothelium growth factor (VEGF), insulin like growth factor‑1 (IGF‑1) and basic fibroblast growth factor (bFGF) in the culture medium were quantified by enzyme‑linked immunosorbent assay. Under both normoxia and physioxia, co‑culture with ASCs protected H2O2‑exposed NRVMs from apoptosis and significantly alleviated the oxidative stress in NRVMs. The protective effects of ASCs were associated with increased secretion of VEGF, IGF‑1 and bFGF. ASCs cultured in 5% O2 exhibited certain cardioprotective effects against H2O2 stress. The results of the present study suggested that O2 concentrations influenced the cardioprotective effects of ASCs. VEGF, IGF‑1 and bFGF may serve a role in the myocardial regeneration mediated by transplanted ASCs.
Collapse
Affiliation(s)
- Yuanyuan Lin
- Department of Cardiology, Shanxi Bethune Hospital, Taiyuan, Shanxi 030032, P.R. China
| | - Xuewen Li
- Department of Cardiology, Shanxi Bethune Hospital, Taiyuan, Shanxi 030032, P.R. China
| | - Chunhui Fan
- Department of Information, Shanxi Bethune Hospital, Taiyuan, Shanxi 030032, P.R. China
| | - Fan Yang
- Department of Cardiology, Shanxi Bethune Hospital, Taiyuan, Shanxi 030032, P.R. China
| | - Dajie Hao
- Department of Cardiology, Shanxi Bethune Hospital, Taiyuan, Shanxi 030032, P.R. China
| | - Wenjia Ge
- Department of Science Research and Education, Shanxi Bethune Hospital, Taiyuan, Shanxi 030032, P.R. China
| | - Yuping Gao
- Department of Cardiology, Shanxi Bethune Hospital, Taiyuan, Shanxi 030032, P.R. China
| |
Collapse
|
26
|
Oliveira BM, Pinto A, Correia A, Ferreira PG, Vilanova M, Teixeira L. Characterization of Myeloid Cellular Populations in Mesenteric and Subcutaneous Adipose Tissue of Holstein-Friesian Cows. Sci Rep 2020; 10:1771. [PMID: 32019985 PMCID: PMC7000716 DOI: 10.1038/s41598-020-58678-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 01/17/2020] [Indexed: 12/12/2022] Open
Abstract
Immune cells resident in adipose tissue have important functions in local and systemic metabolic homeostasis. Nevertheless, these immune cell populations remain poorly characterized in bovines. Recently, we described diverse lymphocyte subpopulations in adipose tissue of Holstein-Friesian cows. Here, we aimed at characterising myeloid cell populations present in bovine adipose tissue using multicolour flow cytometry, cell sorting and histochemistry/immunohistochemistry. Macrophages, CD14+CD11b+MHC-II+CD45+ cells, were identified in mesenteric and subcutaneous adipose tissue, though at higher proportions in the latter. Mast cells, identified as SSC-AhighCD11b−/+CD14−MHC-II−CH138A−CD45+ cells, were also observed in adipose tissue and found at higher proportions than macrophages in mesenteric adipose tissue. Neutrophils, presenting a CH138A+CD11b+ phenotype, were also detected in mesenteric and subcutaneous adipose tissue, however, at much lower frequencies than in the blood. Our gating strategy allowed identification of eosinophils in blood but not in adipose tissue although being detected by morphological analysis at low frequencies in some animals. A population not expressing CD45 and with the CH138A+ CD11b−MHC-II− phenotype, was found abundant and present at higher proportions in mesenteric than subcutaneous adipose tissue. The work reported here may be useful for further studies addressing the function of the described cells.
Collapse
Affiliation(s)
- Bárbara M Oliveira
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.,UMIB - Unidade Multidisciplinar de Investigação Biomédica, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Ana Pinto
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.,UMIB - Unidade Multidisciplinar de Investigação Biomédica, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Alexandra Correia
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,IBMC - Instituto de Biologia Molecular e Celular, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
| | - Paula G Ferreira
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.,UMIB - Unidade Multidisciplinar de Investigação Biomédica, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Manuel Vilanova
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.,I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,IBMC - Instituto de Biologia Molecular e Celular, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
| | - Luzia Teixeira
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal. .,UMIB - Unidade Multidisciplinar de Investigação Biomédica, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.
| |
Collapse
|
27
|
Olivera A, Rivera J. Paradigm Shifts in Mast Cell and Basophil Biology and Function: An Emerging View of Immune Regulation in Health and Disease. Methods Mol Biol 2020; 2163:3-31. [PMID: 32766962 DOI: 10.1007/978-1-0716-0696-4_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The physiological role of the mast cell and basophil has for many years remained enigmatic. In this chapter, we briefly summarize some of the more recent studies that shed new light on the role of mast cells and basophils in health and disease. What we gain from these studies is a new appreciation for mast cells and basophils as sentinels in host defense and a further understanding that dysregulation of mast cell and basophil function can be a component of various diseases other than allergies. Perhaps the most important insight reaped from this work is the increasing awareness that mast cells and basophils can function as immunoregulatory cells that modulate the immune response in health and disease. Collectively, the recent knowledge provides new challenges and opportunities toward the development of novel therapeutic strategies to augment host protection and modify disease through manipulation of mast cell and basophil function.
Collapse
Affiliation(s)
- Ana Olivera
- Molecular Immunology Section, Laboratory of Molecular Immunogenetics, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA.
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, USA.
| | - Juan Rivera
- Molecular Immunology Section, Laboratory of Molecular Immunogenetics, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
28
|
Bourgeois C, Gorwood J, Barrail-Tran A, Lagathu C, Capeau J, Desjardins D, Le Grand R, Damouche A, Béréziat V, Lambotte O. Specific Biological Features of Adipose Tissue, and Their Impact on HIV Persistence. Front Microbiol 2019; 10:2837. [PMID: 31921023 PMCID: PMC6927940 DOI: 10.3389/fmicb.2019.02837] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 11/22/2019] [Indexed: 12/19/2022] Open
Abstract
Although white AT can contribute to anti-infectious immune responses, it can also be targeted and perturbed by pathogens. The AT's immune involvement is primarily due to strong pro-inflammatory responses (with both local and paracrine effects), and the large number of fat-resident macrophages. Adipocytes also exert direct antimicrobial responses. In recent years, it has been found that memory T cells accumulate in AT, where they provide efficient secondary responses against viral pathogens. These observations have prompted researchers to re-evaluate the links between obesity and susceptibility to infections. In contrast, AT serves as a reservoir for several persistence pathogens, such as human adenovirus Ad-36, Trypanosoma gondii, Mycobacterium tuberculosis, influenza A virus, and cytomegalovirus (CMV). The presence and persistence of bacterial DNA in AT has led to the concept of a tissue-specific microbiota. The unexpected coexistence of immune cells and pathogens within the specific AT environment is intriguing, and its impact on anti-infectious immune responses requires further evaluation. AT has been recently identified as a site of HIV persistence. In the context of HIV infection, AT is targeted by both the virus and the antiretroviral drugs. AT's intrinsic metabolic features, large overall mass, and wide distribution make it a major tissue reservoir, and one that may contribute to the pathophysiology of chronic HIV infections. Here, we review the immune, metabolic, viral, and pharmacological aspects that contribute to HIV persistence in AT. We also evaluate the respective impacts of both intrinsic and HIV-induced factors on AT's involvement as a viral reservoir. Lastly, we examine the potential consequences of HIV persistence on the metabolic and immune activities of AT.
Collapse
Affiliation(s)
- Christine Bourgeois
- Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris Sud, INSERM U1184, Fontenay-aux-Roses, France
| | - Jennifer Gorwood
- INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-Métabolisme et Nutrition (ICAN), Sorbonne Université, Paris, France
| | - Aurélie Barrail-Tran
- Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris Sud, INSERM U1184, Fontenay-aux-Roses, France
- AP-HP, Service de Médecine Interne et Immunologie Clinique, Hôpital Bicêtre, Groupe Hospitalier Universitaire Paris Sud, Le Kremlin-Bicêtre, France
| | - Claire Lagathu
- INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-Métabolisme et Nutrition (ICAN), Sorbonne Université, Paris, France
| | - Jacqueline Capeau
- INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-Métabolisme et Nutrition (ICAN), Sorbonne Université, Paris, France
| | - Delphine Desjardins
- Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris Sud, INSERM U1184, Fontenay-aux-Roses, France
| | - Roger Le Grand
- Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris Sud, INSERM U1184, Fontenay-aux-Roses, France
| | - Abderaouf Damouche
- Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris Sud, INSERM U1184, Fontenay-aux-Roses, France
| | - Véronique Béréziat
- INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-Métabolisme et Nutrition (ICAN), Sorbonne Université, Paris, France
| | - Olivier Lambotte
- Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris Sud, INSERM U1184, Fontenay-aux-Roses, France
- AP-HP, Service de Médecine Interne et Immunologie Clinique, Hôpital Bicêtre, Groupe Hospitalier Universitaire Paris Sud, Le Kremlin-Bicêtre, France
| |
Collapse
|
29
|
Zhang D, Dong Y, Zhang Y, Su X, Chen T, Zhang Y, Wu B, Xu G. Spatial distribution and correlation of adipocytes and mast cells in superficial fascia in rats. Histochem Cell Biol 2019; 152:439-451. [DOI: 10.1007/s00418-019-01812-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2019] [Indexed: 12/21/2022]
|
30
|
Varricchi G, de Paulis A, Marone G, Galli SJ. Future Needs in Mast Cell Biology. Int J Mol Sci 2019; 20:E4397. [PMID: 31500217 PMCID: PMC6769913 DOI: 10.3390/ijms20184397] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/02/2019] [Accepted: 09/04/2019] [Indexed: 12/14/2022] Open
Abstract
The pathophysiological roles of mast cells are still not fully understood, over 140 years since their description by Paul Ehrlich in 1878. Initial studies have attempted to identify distinct "subpopulations" of mast cells based on a relatively small number of biochemical characteristics. More recently, "subtypes" of mast cells have been described based on the analysis of transcriptomes of anatomically distinct mouse mast cell populations. Although mast cells can potently alter homeostasis, in certain circumstances, these cells can also contribute to the restoration of homeostasis. Both solid and hematologic tumors are associated with the accumulation of peritumoral and/or intratumoral mast cells, suggesting that these cells can help to promote and/or limit tumorigenesis. We suggest that at least two major subsets of mast cells, MC1 (meaning anti-tumorigenic) and MC2 (meaning pro-tumorigenic), and/or different mast cell mediators derived from otherwise similar cells, could play distinct or even opposite roles in tumorigenesis. Mast cells are also strategically located in the human myocardium, in atherosclerotic plaques, in close proximity to nerves and in the aortic valve. Recent studies have revealed evidence that cardiac mast cells can participate both in physiological and pathological processes in the heart. It seems likely that different subsets of mast cells, like those of cardiac macrophages, can exert distinct, even opposite, effects in different pathophysiological processes in the heart. In this chapter, we have commented on possible future needs of the ongoing efforts to identify the diverse functions of mast cells in health and disease.
Collapse
Affiliation(s)
- Gilda Varricchi
- Department of Translational Medical Sciences (DISMET), University of Naples Federico II, 80138 Naples, Italy.
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, School of Medicine, 80138 Naples, Italy.
- WAO Center of Excellence, 80138 Naples, Italy.
| | - Amato de Paulis
- Department of Translational Medical Sciences (DISMET), University of Naples Federico II, 80138 Naples, Italy.
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, School of Medicine, 80138 Naples, Italy.
- WAO Center of Excellence, 80138 Naples, Italy.
| | - Gianni Marone
- Department of Translational Medical Sciences (DISMET), University of Naples Federico II, 80138 Naples, Italy.
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, School of Medicine, 80138 Naples, Italy.
- WAO Center of Excellence, 80138 Naples, Italy.
- Institute of Experimental Endocrinology and Oncology "Gaetano Salvatore" (IEOS), National Research Council (CNR), 80138 Naples, Italy.
| | - Stephen J Galli
- Departments of Pathology and of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305-5176, USA.
| |
Collapse
|
31
|
Deciphering the differentiation trajectory from hematopoietic stem cells to mast cells. Blood Adv 2019; 2:2273-2281. [PMID: 30206100 DOI: 10.1182/bloodadvances.2018019539] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 08/18/2018] [Indexed: 01/22/2023] Open
Abstract
Hematopoietic stem cells differentiate into all types of blood cells, including peripheral tissue-resident mast cells. The early mast cell differentiation takes place in the bone marrow, after which the progenitor cells enter the circulation and mature once reaching their target organ. Early results from single-cell culture experiments and colony-forming assays have produced the classic hierarchical tree model of hematopoiesis. The introduction of high-throughput, single-cell RNA sequencing is now revolutionizing our understanding of the differentiation process, questioning the classic tree-based models. By integrating the results from early cell culture experiments with single-cell transcriptomics, we present a differentiation landscape model of hematopoiesis and discuss it with focus on mast cells. The review also describes how the hematologic neoplasm systemic mastocytosis can be used to model human hematopoiesis using naturally occurring cell barcoding by means of the common KIT D816V mutation.
Collapse
|
32
|
Méndez-Enríquez E, Hallgren J. Mast Cells and Their Progenitors in Allergic Asthma. Front Immunol 2019; 10:821. [PMID: 31191511 PMCID: PMC6548814 DOI: 10.3389/fimmu.2019.00821] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 03/28/2019] [Indexed: 12/16/2022] Open
Abstract
Mast cells and their mediators have been implicated in the pathogenesis of asthma and allergy for decades. Allergic asthma is a complex chronic lung disease in which several different immune cells, genetic factors and environmental exposures influence the pathology. Mast cells are key players in the asthmatic response through secretion of a multitude of mediators with pro-inflammatory and airway-constrictive effects. Well-known mast cell mediators, such as histamine and bioactive lipids are responsible for many of the physiological effects observed in the acute phase of allergic reactions. The accumulation of mast cells at particular sites of the allergic lung is likely relevant to the asthma phenotype, severity and progression. Mast cells located in different compartments in the lung and airways have different characteristics and express different mediators. According to in vivo experiments in mice, lung mast cells develop from mast cell progenitors induced by inflammatory stimuli to migrate to the airways. Human mast cell progenitors have been identified in the blood circulation. A high frequency of circulating human mast cell progenitors may reflect ongoing pathological changes in the allergic lung. In allergic asthma, mast cells become activated mainly via IgE-mediated crosslinking of the high affinity receptor for IgE (FcεRI) with allergens. However, mast cells can also be activated by numerous other stimuli e.g. toll-like receptors and MAS-related G protein-coupled receptor X2. In this review, we summarize research with implications on the role and development of mast cells and their progenitors in allergic asthma and cover selected activation pathways and mast cell mediators that have been implicated in the pathogenesis. The review places an emphasis on describing mechanisms identified using in vivo mouse models and data obtained by analysis of clinical samples.
Collapse
Affiliation(s)
- Erika Méndez-Enríquez
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Jenny Hallgren
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
33
|
Gurung P, Moussa K, Adams-Huet B, Devaraj S, Jialal I. Increased mast cell abundance in adipose tissue of metabolic syndrome: relevance to the proinflammatory state and increased adipose tissue fibrosis. Am J Physiol Endocrinol Metab 2019; 316:E504-E509. [PMID: 30620639 DOI: 10.1152/ajpendo.00462.2018] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Metabolic Syndrome (MetS) affects 35% of American adults > 40 yr and portends an increased risk for both atherosclerotic cardiovascular disease (ASCVD) and diabetes. The role of mast cells in the proinflammatory state of MetS is not well elucidated. We propose that mast cells in subcutaneous adipose tissue (SAT) of MetS patients without diabetes or clinical ASCVD contribute to insulin resistance and inflammation. Matched controls ( n = 15) and MetS ( n = 19) subjects were recruited from Sacramento, CA, and selected based on Adult Treatment Panel III criteria. SAT biopsy was performed on all subjects and processed for immunohistochemistry. The SAT sections were stained using Astra Blue stain and tryptase stain for mast cells. Fasting blood was obtained for chemistries and biomarkers. Abundance of mast cells (Astra Blue stain) in SAT of MetS subjects compared with controls was increased 2.5-fold ( P < 0.0001). Mast cells correlated positively and significantly with waist circumference, glucose, triglycerides, homeostatic model of assessment-insulin resistance (HOMA-IR), AT insulin resistance, leptin, interleukin (IL)-1β, IL-6, chemerin, p38 MAPK activity, and nuclear factor κB activity in circulating monocytes. Mast cells also correlated significantly with markers of fibrosis and angiogenesis. Tryptase staining of mast cells in AT revealed a significant increase ( P = 0.008) with similar correlations. We make the novel observation that there are increased mast cells in SAT of MetS, and these mast cells correlate with insulin resistance (hepatic and adipose tissue), inflammation, and AT fibrosis. Hence, these immune cells appear to occupy a pivotal role in the pathogenesis of MetS.
Collapse
Affiliation(s)
- Purnima Gurung
- California Northstate University College of Medicine, Section of Endocrinology, Sacramento, California
| | - Karine Moussa
- California Northstate University College of Medicine, Section of Endocrinology, Sacramento, California
| | | | - Sridevi Devaraj
- University of Texas Southwestern Medical Center , Dallas, Texas
| | - Ishwarlal Jialal
- California Northstate University College of Medicine, Section of Endocrinology, Sacramento, California
| |
Collapse
|
34
|
Plotkin JD, Elias MG, Fereydouni M, Daniels-Wells TR, Dellinger AL, Penichet ML, Kepley CL. Human Mast Cells From Adipose Tissue Target and Induce Apoptosis of Breast Cancer Cells. Front Immunol 2019; 10:138. [PMID: 30833944 PMCID: PMC6387946 DOI: 10.3389/fimmu.2019.00138] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 01/16/2019] [Indexed: 01/09/2023] Open
Abstract
Mast cells (MC) are important immune sentinels found in most tissue and widely recognized for their role as mediators of Type I hypersensitivity. However, they also secrete anti-cancer mediators such as tumor necrosis factor alpha (TNF-α) and granulocyte-macrophage colony-stimulating factor (GM-CSF). The purpose of this study was to investigate adipose tissue as a new source of MC in quantities that could be used to study MC biology focusing on their ability to bind to and kill breast cancer cells. We tested several cell culture media previously demonstrated to induce MC differentiation. We report here the generation of functional human MC from adipose tissue. The adipose-derived mast cells (ADMC) are phenotypically and functionally similar to connective tissue expressing tryptase, chymase, c-kit, and FcεRI and capable of degranulating after cross-linking of FcεRI. The ADMC, sensitized with anti-HER2/neu IgE antibodies with human constant regions (trastuzumab IgE and/or C6MH3-B1 IgE), bound to and released MC mediators when incubated with HER2/neu-positive human breast cancer cells (SK-BR-3 and BT-474). Importantly, the HER2/neu IgE-sensitized ADMC induced breast cancer cell (SK-BR-3) death through apoptosis. Breast cancer cell apoptosis was observed after the addition of cell-free supernatants containing mediators released from FcεRI-challenged ADMC. Apoptosis was significantly reduced when TNF-α blocking antibodies were added to the media. Adipose tissue represents a source MC that could be used for multiple research purposes and potentially as a cell-mediated cancer immunotherapy through the expansion of autologous (or allogeneic) MC that can be targeted to tumors through IgE antibodies recognizing tumor specific antigens.
Collapse
Affiliation(s)
- Jesse D Plotkin
- Department of Nanoscience, Nanobiology, Joint School of Nanoscience and Nanoengineering, University of North Carolina, Greensboro, NC, United States
| | - Michael G Elias
- Department of Nanoscience, Nanobiology, Joint School of Nanoscience and Nanoengineering, University of North Carolina, Greensboro, NC, United States
| | - Mohammad Fereydouni
- Department of Nanoscience, Nanobiology, Joint School of Nanoscience and Nanoengineering, University of North Carolina, Greensboro, NC, United States
| | - Tracy R Daniels-Wells
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Anthony L Dellinger
- Department of Nanoscience, Nanobiology, Joint School of Nanoscience and Nanoengineering, University of North Carolina, Greensboro, NC, United States
| | - Manuel L Penichet
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, United States.,The Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States.,AIDS Institute, University of California, Los Angeles, Los Angeles, CA, United States.,The California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, United States
| | - Christopher L Kepley
- Department of Nanoscience, Nanobiology, Joint School of Nanoscience and Nanoengineering, University of North Carolina, Greensboro, NC, United States
| |
Collapse
|
35
|
Hussain M, Bonilla-Rosso G, Kwong Chung CKC, Bäriswyl L, Rodriguez MP, Kim BS, Engel P, Noti M. High dietary fat intake induces a microbiota signature that promotes food allergy. J Allergy Clin Immunol 2019; 144:157-170.e8. [PMID: 30768991 DOI: 10.1016/j.jaci.2019.01.043] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 12/17/2018] [Accepted: 01/23/2019] [Indexed: 01/16/2023]
Abstract
BACKGROUND Diet-induced obesity and food allergies increase in tandem, but a potential cause-and-effect relationship between these diseases of affluence remains to be tested. OBJECTIVE We sought to test the role of high dietary fat intake, diet-induced obesity, and associated changes in gut microbial community structure on food allergy pathogenesis. METHODS Mice were fed a high-fat diet (HFD) for 12 weeks before food allergen sensitization on an atopic dermatitis-like skin lesion, followed by intragastric allergen challenge to induce experimental food allergy. Germ-free animals were colonized with a signature HFD or lean microbiota for 8 weeks before induction of food allergy. Food-induced allergic responses were quantified by using a clinical allergy score, serum IgE levels, serum mouse mast cell protease 1 concentrations, and type 2 cytokine responses. Accumulation of intestinal mast cells was examined by using flow cytometry and chloroacetate esterase tissue staining. Changes in the gut microbial community structure were assessed by using high-throughput 16S ribosomal DNA gene sequencing. RESULTS HFD-induced obesity potentiates food-induced allergic responses associated with dysregulated intestinal effector mast cell responses, increased intestinal permeability, and gut dysbiosis. An HFD-associated microbiome was transmissible to germ-free mice, with the gut microbial community structure of recipients segregating according to the microbiota input source. Independent of an obese state, an HFD-associated gut microbiome was sufficient to confer enhanced susceptibility to food allergy. CONCLUSION These findings identify HFD-induced microbial alterations as risk factors for experimental food allergy and uncouple a pathogenic role of an HFD-associated microbiome from obesity. Postdieting microbiome alterations caused by overindulgence of dietary fat might increase susceptibility to food allergy.
Collapse
Affiliation(s)
- Maryam Hussain
- Institute of Pathology, Department of Experimental Pathology, University of Bern, Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Germán Bonilla-Rosso
- Department of Fundamental Microbiology, University of Lausanne, Lausanne, Switzerland
| | - Cheong K C Kwong Chung
- Institute of Pathology, Department of Experimental Pathology, University of Bern, Bern, Switzerland
| | - Lukas Bäriswyl
- Institute of Pathology, Department of Experimental Pathology, University of Bern, Bern, Switzerland
| | - Maria Pena Rodriguez
- Institute of Pathology, Department of Experimental Pathology, University of Bern, Bern, Switzerland
| | - Brian S Kim
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St Louis, Mo
| | - Philipp Engel
- Department of Fundamental Microbiology, University of Lausanne, Lausanne, Switzerland
| | - Mario Noti
- Institute of Pathology, Department of Experimental Pathology, University of Bern, Bern, Switzerland.
| |
Collapse
|
36
|
Paupert J, Espinosa E, Cenac N, Robert V, Laharrague P, Evrard SM, Casteilla L, Lorsignol A, Cousin B. Rapid and Efficient Production of Human Functional Mast Cells through a Three-Dimensional Culture of Adipose Tissue-Derived Stromal Vascular Cells. THE JOURNAL OF IMMUNOLOGY 2018; 201:3815-3821. [PMID: 30446570 DOI: 10.4049/jimmunol.1701751] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 10/16/2018] [Indexed: 12/26/2022]
Abstract
Mast cells (MC) are innate immune cells involved in many physiological and pathological processes. However, studies of MC function and biology are hampered by the difficulties to obtain human primary MC. To solve this problem, we established a new method to produce easily and rapidly high numbers of MC for in vitro studies using human adipose tissue, which is an abundant and easy access tissue. Stromal vascular fraction of adipose tissue, obtained from human abdominal dermolipectomy, was cultured as spheroids in serum free medium supplemented in stem cell factor. Using this method, we generated, within 3 wk, a highly pure population of connective tissue-type MC expressing MC typical peptidases (tryptase, chymase, and carboxypeptidase-A3) with a yield increasing over time. Stem cell factor was required for this culture, but unlike MC derived from CD34+ cells, this culture did not depend on IL-3 and -6. MC obtained with this method degranulated following FcεRI cross-linking or stimulation by C5a, compound 48/80, and substance P. Interestingly, activation by anti-IgE of both white adipose tissue-MC and MC obtained from peripheral blood-derived CD34+ pluripotent progenitor cells induced the production of PGs as well as proinflammatory cytokines (TNF-α, Il-6, and GM-CSF). In conclusion, we developed a new time saving and reproducible method to produce highly pure and functional human MC in 3 wk from human adipose tissue.
Collapse
Affiliation(s)
- Jenny Paupert
- STROMALab, Université de Toulouse, CNRS 11 Équipe de Recherche Labellisée 5311, Établissement Français du Sang, École Nationale Vétérinaire de Toulouse, INSERM U1031, Université Paul Sabatier, 31100 Toulouse, France.,Université Toulouse III-Université Paul Sabatier, F-31062 Toulouse
| | - Eric Espinosa
- Université Toulouse III-Université Paul Sabatier, F-31062 Toulouse.,INSERM, U1037, Centre de Recherche en Cancérologie de Toulouse, F-31037 Toulouse, France
| | - Nicolas Cenac
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, École Nationale Vétérinaire de Toulouse, Université Paul Sabatier, 31062 Toulouse, France
| | - Virginie Robert
- STROMALab, Université de Toulouse, CNRS 11 Équipe de Recherche Labellisée 5311, Établissement Français du Sang, École Nationale Vétérinaire de Toulouse, INSERM U1031, Université Paul Sabatier, 31100 Toulouse, France.,Université Toulouse III-Université Paul Sabatier, F-31062 Toulouse
| | - Patrick Laharrague
- STROMALab, Université de Toulouse, CNRS 11 Équipe de Recherche Labellisée 5311, Établissement Français du Sang, École Nationale Vétérinaire de Toulouse, INSERM U1031, Université Paul Sabatier, 31100 Toulouse, France.,Université Toulouse III-Université Paul Sabatier, F-31062 Toulouse
| | - Solène M Evrard
- Département d'Histologie et d'Embryologie, École de Médecine de Rangueil, Université Toulouse III-Université Paul Sabatier, 31059 Toulouse Cedex 9, France; and.,Département de Pathologie et Cytologie, Centre Hospitalier Universitaire Toulouse, l'Institut Universitaire du Cancer de Toulouse Oncopole, 31100 Toulouse Cedex 9, France
| | - Louis Casteilla
- STROMALab, Université de Toulouse, CNRS 11 Équipe de Recherche Labellisée 5311, Établissement Français du Sang, École Nationale Vétérinaire de Toulouse, INSERM U1031, Université Paul Sabatier, 31100 Toulouse, France.,Université Toulouse III-Université Paul Sabatier, F-31062 Toulouse
| | - Anne Lorsignol
- STROMALab, Université de Toulouse, CNRS 11 Équipe de Recherche Labellisée 5311, Établissement Français du Sang, École Nationale Vétérinaire de Toulouse, INSERM U1031, Université Paul Sabatier, 31100 Toulouse, France.,Université Toulouse III-Université Paul Sabatier, F-31062 Toulouse
| | - Béatrice Cousin
- STROMALab, Université de Toulouse, CNRS 11 Équipe de Recherche Labellisée 5311, Établissement Français du Sang, École Nationale Vétérinaire de Toulouse, INSERM U1031, Université Paul Sabatier, 31100 Toulouse, France; .,Université Toulouse III-Université Paul Sabatier, F-31062 Toulouse
| |
Collapse
|
37
|
Ford SM, Simon Peter L, Berner P, Cook G, Vande Stouwe C, Dufour J, Bagby G, Nelson S, Molina PE. Differential contribution of chronic binge alcohol and antiretroviral therapy to metabolic dysregulation in SIV-infected male macaques. Am J Physiol Endocrinol Metab 2018; 315:E892-E903. [PMID: 30040479 PMCID: PMC6293168 DOI: 10.1152/ajpendo.00175.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/20/2018] [Accepted: 07/23/2018] [Indexed: 02/08/2023]
Abstract
The incidence of alcohol use disorder (AUD) is higher among people living with HIV (PLWH). The advent and continued development of antiretroviral therapy (ART) has significantly reduced mortality, shifting the course of HIV infection to a chronic illness. However, this is associated with an increased incidence of comorbid conditions, including type 2 diabetes mellitus, insulin resistance, and cardiovascular complications. Using a nonhuman primate model of simian immunodeficiency virus (SIV) infection, previous studies have demonstrated that chronic binge alcohol (CBA) administration decreases whole body insulin responsiveness, irrespective of ART administration. The objective of the current study was to determine the effects of CBA and ART on insulin-sensitive peripheral tissues before the development of overt clinical symptoms of SIV disease. Our results show that CBA reduced omental adipocyte cell size, increased collagen expression, and decreased the in vitro differentiation potential of adipose-derived stem cells. In contrast, it did not alter skeletal muscle or omental or hepatic expression of insulin signaling proteins. However, ART significantly decreased skeletal muscle expression of phosphatase and tensin homolog, total mechanistic target of rapamycin, and ribosomal protein S6. In addition, ART increased hepatic phosphorylation of AMP-activated protein kinase α and increased gene expression of key enzymes required for gluconeogenesis and fatty acid synthesis. These findings suggest that CBA and ART differentially promote adverse metabolic effects in an organ-specific manner that may underlie insulin resistance associated with alcohol, SIV, and ART. Whether this is translated in PLWH with AUD remains to be determined.
Collapse
Affiliation(s)
- Stephen M Ford
- Department of Physiology, Comprehensive Alcohol Research Center, Louisiana State University Health Sciences Center , New Orleans, Louisiana
| | - Liz Simon Peter
- Department of Physiology, Comprehensive Alcohol Research Center, Louisiana State University Health Sciences Center , New Orleans, Louisiana
| | - Paul Berner
- Department of Physiology, Comprehensive Alcohol Research Center, Louisiana State University Health Sciences Center , New Orleans, Louisiana
| | - Garth Cook
- Department of Physiology, Comprehensive Alcohol Research Center, Louisiana State University Health Sciences Center , New Orleans, Louisiana
| | - Curtis Vande Stouwe
- Department of Physiology, Comprehensive Alcohol Research Center, Louisiana State University Health Sciences Center , New Orleans, Louisiana
| | - Jason Dufour
- Divison of Veterinary Medicine, Tulane National Primate Research Center , Covington, Louisiana
| | - Gregory Bagby
- Department of Physiology, Comprehensive Alcohol Research Center, Louisiana State University Health Sciences Center , New Orleans, Louisiana
| | - Steve Nelson
- School of Medicine, Louisiana State University Health Sciences Center , New Orleans, Louisiana
| | - Patricia E Molina
- Department of Physiology, Comprehensive Alcohol Research Center, Louisiana State University Health Sciences Center , New Orleans, Louisiana
| |
Collapse
|
38
|
Abstract
INTRODUCTION There is a major epidemic of obesity, and many obese patients suffer with respiratory symptoms and disease. The overall impact of obesity on lung function is multifactorial, related to mechanical and inflammatory aspects of obesity. Areas covered: Obesity causes substantial changes to the mechanics of the lungs and chest wall, and these mechanical changes cause asthma and asthma-like symptoms such as dyspnea, wheeze, and airway hyperresponsiveness. Excess adiposity is also associated with increased production of inflammatory cytokines and immune cells that may also lead to disease. This article reviews the literature addressing the relationship between obesity and lung function, and studies addressing how the mechanical and inflammatory effects of obesity might lead to changes in lung mechanics and pulmonary function in obese adults and children. Expert commentary: Obesity has significant effects on respiratory function, which contribute significantly to the burden of respiratory disease. These mechanical effects are not readily quantified with conventional pulmonary function testing and measurement of body mass index. Changes in mediators produced by adipose tissue likely also contribute to altered lung function, though as of yet this is poorly understood.
Collapse
Affiliation(s)
- Anne E Dixon
- a Division of Pulmonary and Critical Care Medicine , University of Vermont Larner College of Medicine , Burlington , Vermont , USA
| | - Ubong Peters
- a Division of Pulmonary and Critical Care Medicine , University of Vermont Larner College of Medicine , Burlington , Vermont , USA
| |
Collapse
|
39
|
Priyadarshini S, Pradhan B, Griebel P, Aich P. Cortisol regulates immune and metabolic processes in murine adipocytes and macrophages through HTR2c and HTR5a serotonin receptors. Eur J Cell Biol 2018; 97:483-492. [PMID: 30097291 DOI: 10.1016/j.ejcb.2018.07.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 07/25/2018] [Accepted: 07/26/2018] [Indexed: 12/22/2022] Open
Abstract
Epidemiological studies implicate stress as an important factor contributing to the increasing prevalence of metabolic disorders. Studies have correlated visceral obesity and atherosclerosis with hyper-cortisolemia, a sequela of chronic psychological stress in humans and animals. Although several hormonal markers of stress have been associated with various metabolic disorders, the mechanism by which these hormones alter metabolic functions have not been established. We used an in vitro model system, culturing 3T3-L1 pre-adipocytes and RAW 264.7 macrophages in the presence or absence of cortisol, to analyze cell signaling pathways mediating changes in metabolic functions. Our analysis revealed that cortisol up-regulated the expression and function of two serotonin (S) receptors, HTR2c and HTR5a. HTR2c and HTR5a were also directly involved in mediating cortisol enhanced adipogenesis when pre-adipocytes were cultured alone or in the presence of macrophages. Finally, cortisol treatment of pre-adipocytes co-cultured with macrophages enhanced adipogenesis in both macrophages and pre-adipocytes.
Collapse
Affiliation(s)
- Sushri Priyadarshini
- School of Biological Sciences, National Institute of Science Education and Research (NISER), HBNI, PO- Bhimpur-Padanpur, Jatni, Khurda, Odisha, 752050, India
| | - Biswaranjan Pradhan
- School of Biological Sciences, National Institute of Science Education and Research (NISER), HBNI, PO- Bhimpur-Padanpur, Jatni, Khurda, Odisha, 752050, India
| | - Philip Griebel
- VIDO-Intervac, University of Saskatchewan, Saskatoon, SK, S7N 5E3, Canada; School of Public Health, University of Saskatchewan, Saskatoon, SK, S7N 5A8, Canada
| | - Palok Aich
- School of Biological Sciences, National Institute of Science Education and Research (NISER), HBNI, PO- Bhimpur-Padanpur, Jatni, Khurda, Odisha, 752050, India.
| |
Collapse
|
40
|
Yang HW, Liu XY, Shen ZF, Yao W, Gong XB, Huang HX, Ding GH. An investigation of the distribution and location of mast cells affected by the stiffness of substrates as a mechanical niche. Int J Biol Sci 2018; 14:1142-1152. [PMID: 29989093 PMCID: PMC6036734 DOI: 10.7150/ijbs.26738] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 06/04/2018] [Indexed: 12/27/2022] Open
Abstract
The distribution and location of mast cells are closely related to their physiological and pathological functions, such as allergic responses, immunity, and fibrosis, and are used in acupuncture. In this study, the distribution of mast cells in vivo was observed, and mechanical clues for understanding their distribution based on mechanical niches were explored. By toluidine blue staining and immunohistochemical staining, we examined the distribution and location of mast cells in rat skin and found that mast cells are distributed in a spatially nonuniform manner, preferring to locate at regions in the tissue and extracellular matrix with stiffness changes. In vitro experiments for studying the distribution of rat basophilic leukemia (RBL-2H3) mast cell line on poly-di-methyl-siloxane (PDMS) substrates with stiffness variations were performed. It was found that RBL-2H3 cells migrate and tend to remain in the areas with stiffness variations. The present research suggests that changing the stiffness of local tissues may stimulate mast cell recruitment, which may be the method by which some traditional Chinese medicine treatments, such as acupuncture. On the basis of the origin of mast cells and our experimental results, we predict that mast cells exist in tissues that contain permeable capillaries and prefer regions with stiffness changes. We discussed this prediction using examples of specific tissues from some cases.
Collapse
Affiliation(s)
- Hong-Wei Yang
- Department of Aeronautics and Astronautics, Fudan University, Shanghai 200433, China
| | - Xin-Yue Liu
- Key Laboratory of Hydrodynamics (Ministry of Education), Department of Engineering Mechanics, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhou-Feng Shen
- Department of Aeronautics and Astronautics, Fudan University, Shanghai 200433, China
| | - Wei Yao
- Department of Aeronautics and Astronautics, Fudan University, Shanghai 200433, China
| | - Xiao-Bo Gong
- Key Laboratory of Hydrodynamics (Ministry of Education), Department of Engineering Mechanics, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hua-Xiong Huang
- Department of Mathematics and Statistics, York University, Toronto, Ontario, Canada, M3J 1P3
| | - Guang-Hong Ding
- Department of Aeronautics and Astronautics, Fudan University, Shanghai 200433, China
| |
Collapse
|
41
|
Żelechowska P, Agier J, Kozłowska E, Brzezińska-Błaszczyk E. Mast cells participate in chronic low-grade inflammation within adipose tissue. Obes Rev 2018; 19:686-697. [PMID: 29334696 DOI: 10.1111/obr.12670] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 11/29/2017] [Accepted: 12/15/2017] [Indexed: 12/26/2022]
Abstract
Obesity is reckoned as one of the civilization diseases, posing a considerable global health issue. Evidence points towards a contribution of multitude immune cell populations in obesity pathomechanism and the development of chronic low-grade inflammation in the expanded adipose tissue. Notably, adipose tissue is a reservoir of mast cells which number in individuals with obesity particularly increased. Some of them tend to degranulation what generate secretion of strong pro-inflammatory and regulatory mediators, as well as cytokines/chemokines. Several lines of evidence suggest that mast cells are strictly associated with pro-inflammatory status in adipose tissue by their indirect impact on immune cell attraction and activation. Furthermore, mast cells affect adipose tissue remodelling and fibrosis by adipocyte differentiation, fibroblast proliferation and enhancing extracellular matrix proteins expression. This review will summarize current knowledge on mast cell features and their role in the development of chronic low-grade inflammation within adipose tissue.
Collapse
Affiliation(s)
- P Żelechowska
- Department of Experimental Immunology, Medical University of Lodz, Lodz, Poland
| | - J Agier
- Department of Experimental Immunology, Medical University of Lodz, Lodz, Poland
| | - E Kozłowska
- Department of Experimental Immunology, Medical University of Lodz, Lodz, Poland
| | | |
Collapse
|
42
|
Muñoz MF, Argüelles S, Guzman-Chozas M, Guillén-Sanz R, Franco JM, Pintor-Toro JA, Cano M, Ayala A. Cell tracking, survival, and differentiation capacity of adipose-derived stem cells after engraftment in rat tissue. J Cell Physiol 2018; 233:6317-6328. [PMID: 29319169 DOI: 10.1002/jcp.26439] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 01/05/2018] [Indexed: 12/30/2022]
Abstract
Adipose tissue is an important source of adipose derived stem cells (ADSCs). These cells have the potential of being used for certain therapies, in which the main objective is to recover the function of a tissue/organ affected by a disease. In order to contribute to repair of the tissue, these cells should be able to survive and carry out their functions in unfavorable conditions after being transplanted. This process requires a better understanding of the biology involved: such as the time cells remain in the implant site, how long they stay there, and whether or not they differentiate into host tissue cells. This report focuses on these questions. ADSC were injected into three different tissues (substantia nigra, ventricle, liver) and they were tracked in vivo with a dual GFP-Luc reporter system. The results show that ADSCs were able to survive up to 4 months after the engraftment and some of them started showing resident cell tissue phenotype. These results demonstrate their long-term capacity of survival and differentiation when injected in vivo.
Collapse
Affiliation(s)
- Mario F Muñoz
- Departamento de Bioquímica y Biología Molecular, Universidad de Sevilla, Sevilla, Spain
| | - Sandro Argüelles
- Departamento de Fisiología, Universidad de Sevilla, Sevilla, Spain
| | - Matias Guzman-Chozas
- Departamento de Nutrición, Bromatología, Toxicología y Medicina Legal, . Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
| | - Remedios Guillén-Sanz
- Departamento de Nutrición, Bromatología, Toxicología y Medicina Legal, . Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
| | - Jaime M Franco
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Departamento de Señalización Celular, Universidad de Sevilla, Sevilla, Spain
| | - José A Pintor-Toro
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Departamento de Señalización Celular, Universidad de Sevilla, Sevilla, Spain
| | - Mercedes Cano
- Departamento de Fisiología, Universidad de Sevilla, Sevilla, Spain
| | - Antonio Ayala
- Departamento de Bioquímica y Biología Molecular, Universidad de Sevilla, Sevilla, Spain
| |
Collapse
|
43
|
Abstract
Solid tumor growth and metastasis require the interaction of tumor cells with the surrounding tissue, leading to a view of tumors as tissue-level phenomena rather than exclusively cell-intrinsic anomalies. Due to the ubiquitous nature of adipose tissue, many types of solid tumors grow in proximate or direct contact with adipocytes and adipose-associated stromal and vascular components, such as fibroblasts and other connective tissue cells, stem and progenitor cells, endothelial cells, innate and adaptive immune cells, and extracellular signaling and matrix components. Excess adiposity in obesity both increases risk of cancer development and negatively influences prognosis in several cancer types, in part due to interaction with adipose tissue cell populations. Herein, we review the cellular and noncellular constituents of the adipose "organ," and discuss the mechanisms by which these varied microenvironmental components contribute to tumor development, with special emphasis on obesity. Due to the prevalence of breast and prostate cancers in the United States, their close anatomical proximity to adipose tissue depots, and their complex epidemiologic associations with obesity, we particularly highlight research addressing the contribution of adipose tissue to the initiation and progression of these cancer types. Obesity dramatically modifies the adipose tissue microenvironment in numerous ways, including induction of fibrosis and angiogenesis, increased stem cell abundance, and expansion of proinflammatory immune cells. As many of these changes also resemble shifts observed within the tumor microenvironment, proximity to adipose tissue may present a hospitable environment to developing tumors, providing a critical link between adiposity and tumorigenesis. © 2018 American Physiological Society. Compr Physiol 8:237-282, 2018.
Collapse
Affiliation(s)
- Alyssa J. Cozzo
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ashley M. Fuller
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Liza Makowski
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
44
|
Cildir G, Pant H, Lopez AF, Tergaonkar V. The transcriptional program, functional heterogeneity, and clinical targeting of mast cells. J Exp Med 2017; 214:2491-2506. [PMID: 28811324 PMCID: PMC5584128 DOI: 10.1084/jem.20170910] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 06/28/2017] [Accepted: 07/26/2017] [Indexed: 12/11/2022] Open
Abstract
Cildir et al. discuss the recent findings in transcriptional regulation of mast cell development and activation and provide insights into the plasticity and clinical targeting of mast cell functions. Mast cells are unique tissue-resident immune cells that express an array of receptors that can be activated by several extracellular cues, including antigen–immunoglobulin E (IgE) complexes, bacteria, viruses, cytokines, hormones, peptides, and drugs. Mast cells constitute a small population in tissues, but their extraordinary ability to respond rapidly by releasing granule-stored and newly made mediators underpins their importance in health and disease. In this review, we document the biology of mast cells and introduce new concepts and opinions regarding their role in human diseases beyond IgE-mediated allergic responses and antiparasitic functions. We bring to light recent discoveries and developments in mast cell research, including regulation of mast cell functions, differentiation, survival, and novel mouse models. Finally, we highlight the current and future opportunities for therapeutic intervention of mast cell functions in inflammatory diseases.
Collapse
Affiliation(s)
- Gökhan Cildir
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia
| | - Harshita Pant
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia.,School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - Angel F Lopez
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia
| | - Vinay Tergaonkar
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia .,Laboratory of NF-κB Signalling, Institute of Molecular and Cell Biology (IMCB), Singapore, Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, Singapore
| |
Collapse
|
45
|
Ngkelo A, Richart A, Kirk JA, Bonnin P, Vilar J, Lemitre M, Marck P, Branchereau M, Le Gall S, Renault N, Guerin C, Ranek MJ, Kervadec A, Danelli L, Gautier G, Blank U, Launay P, Camerer E, Bruneval P, Menasche P, Heymes C, Luche E, Casteilla L, Cousin B, Rodewald HR, Kass DA, Silvestre JS. Mast cells regulate myofilament calcium sensitization and heart function after myocardial infarction. J Exp Med 2017; 213:1353-74. [PMID: 27353089 PMCID: PMC4925026 DOI: 10.1084/jem.20160081] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 05/12/2016] [Indexed: 11/24/2022] Open
Abstract
Ngkelo et al. use a mast cell–deficient mouse model to reveal a protective role of mast cells in myocardial infarction, through regulation of the cardiac contractile machinery. Acute myocardial infarction (MI) is a severe ischemic disease responsible for heart failure and sudden death. Inflammatory cells orchestrate postischemic cardiac remodeling after MI. Studies using mice with defective mast/stem cell growth factor receptor c-Kit have suggested key roles for mast cells (MCs) in postischemic cardiac remodeling. Because c-Kit mutations affect multiple cell types of both immune and nonimmune origin, we addressed the impact of MCs on cardiac function after MI, using the c-Kit–independent MC-deficient (Cpa3Cre/+) mice. In response to MI, MC progenitors originated primarily from white adipose tissue, infiltrated the heart, and differentiated into mature MCs. MC deficiency led to reduced postischemic cardiac function and depressed cardiomyocyte contractility caused by myofilament Ca2+ desensitization. This effect correlated with increased protein kinase A (PKA) activity and hyperphosphorylation of its targets, troponin I and myosin-binding protein C. MC-specific tryptase was identified to regulate PKA activity in cardiomyocytes via protease-activated receptor 2 proteolysis. This work reveals a novel function for cardiac MCs modulating cardiomyocyte contractility via alteration of PKA-regulated force–Ca2+ interactions in response to MI. Identification of this MC-cardiomyocyte cross-talk provides new insights on the cellular and molecular mechanisms regulating the cardiac contractile machinery and a novel platform for therapeutically addressable regulators.
Collapse
Affiliation(s)
- Anta Ngkelo
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France
| | - Adèle Richart
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France
| | - Jonathan A Kirk
- Division of Cardiology, Johns Hopkins Medical Institutions, Baltimore, MD 212015
| | - Philippe Bonnin
- INSERM, U965, Hôpital Lariboisière-Fernand-Widal, Assistance Publique Hôpitaux de Paris, F-75010 Paris, France
| | - Jose Vilar
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France
| | - Mathilde Lemitre
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France
| | - Pauline Marck
- INSERM, UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, F-31004 Toulouse, France
| | - Maxime Branchereau
- INSERM, UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, F-31004 Toulouse, France
| | - Sylvain Le Gall
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France
| | - Nisa Renault
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France
| | - Coralie Guerin
- National Cytometry Platform, Department of Infection and Immunity, Luxembourg Institute of Health, L-4354 Esch-sur-Alzette, Luxembourg
| | - Mark J Ranek
- Division of Cardiology, Johns Hopkins Medical Institutions, Baltimore, MD 212015
| | - Anaïs Kervadec
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France
| | - Luca Danelli
- Laboratoire d'Excellence INFLAMEX, Université Paris Diderot, Sorbonne Paris Cité, F-75018 Paris, France INSERM, U1149, F-75018 Paris, France Centre National de la Recherche Scientifique (CNRS) ERL 8252, F-75018 Paris, France
| | - Gregory Gautier
- Laboratoire d'Excellence INFLAMEX, Université Paris Diderot, Sorbonne Paris Cité, F-75018 Paris, France INSERM, U1149, F-75018 Paris, France
| | - Ulrich Blank
- Laboratoire d'Excellence INFLAMEX, Université Paris Diderot, Sorbonne Paris Cité, F-75018 Paris, France INSERM, U1149, F-75018 Paris, France Centre National de la Recherche Scientifique (CNRS) ERL 8252, F-75018 Paris, France
| | - Pierre Launay
- Laboratoire d'Excellence INFLAMEX, Université Paris Diderot, Sorbonne Paris Cité, F-75018 Paris, France INSERM, U1149, F-75018 Paris, France
| | - Eric Camerer
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France
| | - Patrick Bruneval
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France Hôpital European George Pompidou, Assistance Publique Hôpitaux de Paris, F-75015 Paris, France
| | - Philippe Menasche
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France Hôpital European George Pompidou, Assistance Publique Hôpitaux de Paris, F-75015 Paris, France
| | - Christophe Heymes
- INSERM, UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, F-31004 Toulouse, France
| | - Elodie Luche
- STROMALab, Etablissement Français du Sang, INSERM U1031, CNRS ERL 5311, Université de Toulouse, F-31004 Toulouse, France
| | - Louis Casteilla
- STROMALab, Etablissement Français du Sang, INSERM U1031, CNRS ERL 5311, Université de Toulouse, F-31004 Toulouse, France
| | - Béatrice Cousin
- STROMALab, Etablissement Français du Sang, INSERM U1031, CNRS ERL 5311, Université de Toulouse, F-31004 Toulouse, France
| | - Hans-Reimer Rodewald
- Division of Cellular Immunology, German Cancer Research Center, D-69120 Heidelberg, Germany
| | - David A Kass
- Division of Cardiology, Johns Hopkins Medical Institutions, Baltimore, MD 212015
| | - Jean-Sébastien Silvestre
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Centre de Recherche Cardiovasculaire, Université Paris Descartes, Sorbonne Paris Cité, F-75015 Paris, France
| |
Collapse
|
46
|
Luche E, Robert V, Cuminetti V, Pomié C, Sastourné-Arrey Q, Waget A, Arnaud E, Varin A, Labit E, Laharrague P, Burcelin R, Casteilla L, Cousin B. Corrupted adipose tissue endogenous myelopoiesis initiates diet-induced metabolic disease. eLife 2017; 6. [PMID: 28656887 PMCID: PMC5509432 DOI: 10.7554/elife.23194] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 06/28/2017] [Indexed: 12/21/2022] Open
Abstract
Activation and increased numbers of inflammatory macrophages, in adipose tissue (AT) are deleterious in metabolic diseases. Up to now, AT macrophages (ATM) accumulation was considered to be due to blood infiltration or local proliferation, although the presence of resident hematopoietic stem/progenitor cells (Lin-/Sca+/c-Kit+; LSK phenotype) in the AT (AT-LSK) has been reported. By using transplantation of sorted AT-LSK and gain and loss of function studies we show that some of the inflammatory ATM inducing metabolic disease, originate from resident AT-LSK. Transplantation of AT-LSK sorted from high fat diet-fed (HFD) mice is sufficient to induce ATM accumulation, and to transfer metabolic disease in control mice. Conversely, the transplantation of control AT-LSK improves both AT-inflammation and glucose homeostasis in HFD mice. Our results clearly demonstrate that resident AT-LSK are one of the key point of metabolic disease, and could thus constitute a new promising therapeutic target to fight against metabolic disease. DOI:http://dx.doi.org/10.7554/eLife.23194.001
Collapse
Affiliation(s)
- Elodie Luche
- STROMALab, Université de Toulouse, CNRS ERL 5311, EFS, INP-ENVT, Inserm U1031, UPS, Toulouse, France
| | - Virginie Robert
- STROMALab, Université de Toulouse, CNRS ERL 5311, EFS, INP-ENVT, Inserm U1031, UPS, Toulouse, France
| | - Vincent Cuminetti
- STROMALab, Université de Toulouse, CNRS ERL 5311, EFS, INP-ENVT, Inserm U1031, UPS, Toulouse, France
| | - Celine Pomié
- INSERM U1048, Université de Toulouse, EFS, INP-ENVT, Inserm U1031, UPS, Toulouse, France
| | - Quentin Sastourné-Arrey
- STROMALab, Université de Toulouse, CNRS ERL 5311, EFS, INP-ENVT, Inserm U1031, UPS, Toulouse, France
| | - Aurélie Waget
- INSERM U1048, Université de Toulouse, EFS, INP-ENVT, Inserm U1031, UPS, Toulouse, France
| | - Emmanuelle Arnaud
- STROMALab, Université de Toulouse, CNRS ERL 5311, EFS, INP-ENVT, Inserm U1031, UPS, Toulouse, France
| | - Audrey Varin
- STROMALab, Université de Toulouse, EFS, INP-ENVT, Inserm U1031, UPS, Toulouse, France
| | - Elodie Labit
- STROMALab, Université de Toulouse, CNRS ERL 5311, EFS, INP-ENVT, Inserm U1031, UPS, Toulouse, France
| | - Patrick Laharrague
- STROMALab, Université de Toulouse, CNRS ERL 5311, EFS, INP-ENVT, Inserm U1031, UPS, Toulouse, France
| | - Remy Burcelin
- INSERM U1048, Université de Toulouse, EFS, INP-ENVT, Inserm U1031, UPS, Toulouse, France
| | - Louis Casteilla
- STROMALab, Université de Toulouse, CNRS ERL 5311, EFS, INP-ENVT, Inserm U1031, UPS, Toulouse, France
| | - Beatrice Cousin
- STROMALab, Université de Toulouse, CNRS ERL 5311, EFS, INP-ENVT, Inserm U1031, UPS, Toulouse, France
| |
Collapse
|
47
|
The Innate Immune Response in Myocardial Infarction, Repair, and Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1003:251-272. [PMID: 28667562 DOI: 10.1007/978-3-319-57613-8_12] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Following myocardial infarction (MI), resident innate immune cells such as macrophages, innate lymphoid cells, and mast cells rapidly coordinate their function to contain inflammation by removing dying cells and promoting cardiomyocyte replenishment. To sustain local tissue repair functions, hematopoietic progenitors are mobilized from the bone marrow to the spleen to generate subsequent myeloid cells such as monocytes and neutrophils, which are rapidly recruited at the site of MI. A finely tuned balance between local adaptation and recruitment controls the overall outcome of the cardiac tissue regeneration versus repair and scar formation.In this chapter, the (potential) roles of the innate immune system residing in the heart are discussed in the context of recent findings about macrophage ontogeny and their homeostasis with circulating monocytes during cardiac tissue growth and after myocardial infarction. Their interactions with other members of the innate immune system are also discussed with a particular emphasis on the potential involvement of mast cells and innate lymphoid cells during MI, largely underestimated until recently. Understanding the development and the functions of the different protagonists responding to MI as well as their potential cross talk could help design new strategies for regenerative medicine intervention.
Collapse
|
48
|
Mast cell activation disease and the modern epidemic of chronic inflammatory disease. Transl Res 2016; 174:33-59. [PMID: 26850903 DOI: 10.1016/j.trsl.2016.01.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Revised: 01/07/2016] [Accepted: 01/08/2016] [Indexed: 12/18/2022]
Abstract
A large and growing portion of the human population, especially in developed countries, suffers 1 or more chronic, often quite burdensome ailments which either are overtly inflammatory in nature or are suspected to be of inflammatory origin, but for which investigations to date have failed to identify specific causes, let alone unifying mechanisms underlying the multiple such ailments that often afflict such patients. Relatively recently described as a non-neoplastic cousin of the rare hematologic disease mastocytosis, mast cell (MC) activation syndrome-suspected to be of greatly heterogeneous, complex acquired clonality in many cases-is a potential underlying/unifying explanation for a diverse assortment of inflammatory ailments. A brief review of MC biology and how aberrant primary MC activation might lead to such a vast range of illness is presented.
Collapse
|
49
|
Lack of Platelet-Activating Factor Receptor Attenuates Experimental Food Allergy but Not Its Metabolic Alterations regarding Adipokine Levels. BIOMED RESEARCH INTERNATIONAL 2016; 2016:8601359. [PMID: 27314042 PMCID: PMC4897668 DOI: 10.1155/2016/8601359] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 04/14/2016] [Accepted: 05/04/2016] [Indexed: 11/17/2022]
Abstract
Platelet-activating factor (PAF) is known to be an important mediator of anaphylaxis. However, there is a lack of information in the literature about the role of PAF in food allergy. The aim of this work was to elucidate the participation of PAF during food allergy development and the consequent adipose tissue inflammation along with its alterations. Our data demonstrated that, both before oral challenge and after 7 days receiving ovalbumin (OVA) diet, OVA-sensitized mice lacking the PAF receptor (PAFR) showed a decreased level of anti-OVA IgE associated with attenuated allergic markers in comparison to wild type (WT) mice. Moreover, there was less body weight and adipose tissue loss in PAFR-deficient mice. However, some features of inflamed adipose tissue presented by sensitized PAFR-deficient and WT mice after oral challenge were similar, such as a higher rate of rolling leukocytes in this tissue and lower circulating levels of adipokines (resistin and adiponectin) in comparison to nonsensitized mice. Therefore, PAF signaling through PAFR is important for the allergic response to OVA but not for the adipokine alterations caused by this inflammatory process. Our work clarifies some effects of PAF during food allergy along with its role on the metabolic consequences of this inflammatory process.
Collapse
|
50
|
Abstract
Adipose tissue has traditionally been defined as connective tissue that stores excess calories in the form of triacylglycerol. However, the physiologic functions attributed to adipose tissue are expanding, and it is now well established that adipose tissue is an endocrine gland. Among the endocrine factors elaborated by adipose tissue are the adipokines; hormones, similar in structure to cytokines, produced by adipose tissue in response to changes in adipocyte triacylglycerol storage and local and systemic inflammation. They inform the host regarding long-term energy storage and have a profound influence on reproductive function, blood pressure regulation, energy homeostasis, the immune response, and many other physiologic processes. The adipokines possess pro- and anti-inflammatory properties and play a critical role in integrating systemic metabolism with immune function. In calorie restriction and starvation, proinflammatory adipokines decline and anti-inflammatory adipokines increase, which informs the host of energy deficits and contributes to the suppression of immune function. In individuals with normal metabolic status, there is a balance of pro- and anti-inflammatory adipokines. This balance shifts to favor proinflammatory mediators as adipose tissue expands during the development of obesity. As a consequence, the proinflammatory status of adipose tissue contributes to a chronic low-grade state of inflammation and metabolic disorders associated with obesity. These disturbances are associated with an increased risk of metabolic disease, type 2 diabetes, cardiovascular disease, and many other pathological conditions. This review focuses on the impact of energy homeostasis on the adipokines in immune function.
Collapse
Affiliation(s)
- Peter Mancuso
- Department of Nutritional Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|