1
|
Wang H, Chen G, Gong Q, Wu J, Chen P. Systemic inflammatory regulators are associated with two common types of neuropathic pain: A bidirectional Mendelian randomization study. Int Immunopharmacol 2024; 143:113466. [PMID: 39471697 DOI: 10.1016/j.intimp.2024.113466] [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: 01/31/2024] [Revised: 09/03/2024] [Accepted: 10/19/2024] [Indexed: 11/01/2024]
Abstract
BACKGROUND Currently, there is limited and inconsistent evidence regarding the causal relationship between systemic inflammatory regulators and two common types of neuropathic pain, namely, postherpetic neuralgia (PHN) and trigeminal neuralgia (TN). This study employed a Mendelian randomization (MR) approach to investigate the causal relationship between systemic inflammatory regulators and these two common neuropathic pain conditions. METHODS In this study, 41 single-nucleotide polymorphisms (SNPs) associated with PHN and TN were selected as instrumental variables (IVs) representing systemic inflammatory regulators. Genetic associations of systemic inflammatory regulators were derived from recent genome-wide association studies (GWAS) on the human proteome and cytokines. Genetic data related to PHN and TN were obtained from the FinnGen. The primary analytical method utilized inverse variance weighting (IVW) and various sensitivity analyses. RESULTS Prior to applying the false discovery rate (FDR) correction, our bidirectional MR analysis revealed that increased levels of IFNγ (OR: 0.46, 95% CI: 0.24-0.87, PIVW: 0.016) and MCP3 (OR: 0.52, 95% CI: 0.35-0.77, PIVW: 0.001) were associated with a reduced risk of PHN, and increased levels of IL-16 (OR: 0.81, 95% CI: 0.67-0.98, PIVW: 0.026) were causally associated with a reduced risk of TN. In discussing the impact of PHN and TN on systemic inflammatory regulator levels, we observed the following findings: The BETA for CTACK was -0.07 (95% CI: -0.13 to -0.01, PIVW: 0.015), the BETA for FGFBasic was -0.04 (95% CI: -0.08 to -0.01, PIVW: 0.020), and the BETA for IL-17 was -0.04 (95% CI: -0.08 to -0.01, PIVW: 0.019). These results indicate that patients with PHN tend to have lower levels of CTACK, FGFBasic, and IL-17. Conversely, the BETA for IFNγ was -0.09 (95% CI: -0.18 to 0.00, PIVW: 0.046), suggesting that patients with TN tend to have lower levels of IFN γ. However, after FDR correction, only the association between MCP3 and PHN remained statistically significant (PFDR: 0.044). CONCLUSION This study found that certain systemic inflammatory regulators are associated with PHN and TN to some extent. However, further research is needed to explore the specific mechanisms underlying these connections.
Collapse
Affiliation(s)
- Hao Wang
- Basic Medical School, Guizhou University of Traditional Chinese Medicine, Guizhou 550025, China
| | - Guanglei Chen
- Basic Medical School, Guizhou University of Traditional Chinese Medicine, Guizhou 550025, China
| | - Qian Gong
- First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jing Wu
- Basic Medical School, Guizhou University of Traditional Chinese Medicine, Guizhou 550025, China
| | - Peng Chen
- Basic Medical School, Guizhou University of Traditional Chinese Medicine, Guizhou 550025, China.
| |
Collapse
|
2
|
Tan ESJ, Choi H, DeFilippi CR, Oon YY, Chan SP, Gong L, Lunaria JB, Liew OW, Chong JPC, Tay ELW, Soo WM, Yip JWL, Yong QW, Lee EM, Daniel Yeo PS, Ding ZP, Tang HC, Ewe SH, Chin CWL, Chai SC, Goh PP, Ling LF, Ong HY, Richards AM, Ling LH. Circulating Plasma Proteins in Aortic Stenosis: Associations With Severity, Myocardial Response, and Clinical Outcomes. J Am Heart Assoc 2024; 13:e035486. [PMID: 39344657 DOI: 10.1161/jaha.124.035486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 08/19/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Echocardiographic indexes of aortic stenosis may not comprehensively reflect disease morbidity. Plasma proteomic profiling may add prognostic value in these patients. METHODS AND RESULTS Proximity extension assays (Olink) of 183 circulating cardiovascular and inflammatory proteins were performed in a prospective follow-up study of 122 asymptomatic/minimally symptomatic patients (mean±SD age, 69.1±10.9 years; 61% men) with moderate to severe aortic stenosis and preserved left ventricular ejection fraction. Protein signatures of higher-risk echocardiographic subgroups were determined. Associations of proteins with the primary composite outcome (heart failure hospitalization, progression to New York Heart Association class III-IV, or all-cause mortality) were evaluated using competing risk analyses, with aortic valve replacement being the competing risk. Network analysis unveiled mutually exclusive communities of proteins and echocardiographic parameters, connected only through NT-proBNP (N-terminal pro-B-type natriuretic peptide). Members of the tumor necrosis factor receptor superfamily (TNFRSF1A, TNFRSF1B, and TNFRSF14), and trefoil factor-3 were major hub proteins among the circulating biomarkers. Left ventricular global longitudinal strain >-15% was associated with higher levels of proteins, primarily of inflammation and immune regulation, whereas aortic valve area <1 cm2, E/e' >15, and left atrial reservoir strain <20% were associated with higher levels of NT-proBNP. Of 14 proteins associated with the primary end point, phospholipase-C, C-X-C motif chemokine-9, and interleukin-10 receptor subunit β demonstrated the highest hazard ratios after adjusting for clinical factors (q<0.05). CONCLUSIONS Plasma proteins involved in inflammation and immune regulation were differentially expressed in patients with aortic stenosis with reduced left ventricular global longitudinal strain, and associated with adverse clinical outcomes. Their incorporation into aortic stenosis risk stratification warrants further assessment.
Collapse
Affiliation(s)
- Eugene S J Tan
- National University Heart Centre Singapore Singapore
- Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
| | - Hyungwon Choi
- Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
- Cardiovascular Research Institute, National University Health System Singapore Singapore
| | | | - Yen-Yee Oon
- Sarawak Heart Centre Kota Samarahan Sarawak Malaysia
| | - Siew-Pang Chan
- National University Heart Centre Singapore Singapore
- Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
| | - Lingli Gong
- Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
| | - Josephine B Lunaria
- Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
| | - Oi-Wah Liew
- Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
- Cardiovascular Research Institute, National University Health System Singapore Singapore
| | - Jenny Pek-Ching Chong
- Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
- Cardiovascular Research Institute, National University Health System Singapore Singapore
| | - Edgar Lik-Wui Tay
- National University Heart Centre Singapore Singapore
- Asian Heart and Vascular Centre Singapore Singapore
| | - Wern-Miin Soo
- National University Heart Centre Singapore Singapore
| | - James Wei-Luen Yip
- National University Heart Centre Singapore Singapore
- Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
| | | | | | - Poh Shuan Daniel Yeo
- Tan Tock Seng Hospital Singapore Singapore
- Apex Heart Clinic Gleneagles Hospital Singapore Singapore
| | | | | | | | | | | | | | | | | | - A Mark Richards
- National University Heart Centre Singapore Singapore
- Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
- Cardiovascular Research Institute, National University Health System Singapore Singapore
- Christchurch Heart Institute, University of Otago Christchurch New Zealand
| | - Lieng-Hsi Ling
- National University Heart Centre Singapore Singapore
- Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
- Cardiovascular Research Institute, National University Health System Singapore Singapore
| |
Collapse
|
3
|
Han Z, Chen Y, Ye X. The causality between smoking and intervertebral disc degeneration mediated by IL-1β secreted by macrophage: A Mendelian randomization study. Heliyon 2024; 10:e37044. [PMID: 39286222 PMCID: PMC11402911 DOI: 10.1016/j.heliyon.2024.e37044] [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: 02/05/2024] [Accepted: 08/27/2024] [Indexed: 09/19/2024] Open
Abstract
There is still a lack of high-level evidence regarding the causal relationship between smoking and intervertebral disc degenerative diseases. This study utilized data from genome wide analysis studies and conducted two-sample Mendelian randomization analyses across multiple heterogeneous datasets. We evaluated the causal relationships between smoking behavior, serum inflammatory factors, serum chemokines, and intervertebral disc degeneration. Sensitivity analysis was performed to examine data heterogeneity and the pleiotropy of causal effects. The results indicated that smokers were liable to develop intervertebral disc degeneration (OR 1.770; 95 % CI, 1.519-2.064; p = 2.992 × 10-13), and long-term smoking behavior increased the risk of intervertebral disc degeneration (OR 1.715; 95 % CI 1.475-1.994; P = 2.220 × 10-12). Additionally, a causal relationship was confirmed between serum IL-1β level and intervertebral disc degeneration (OR 1.087; 95 % CI, 1.023-1.154; p = 0.007). The "smoking index" representing lifelong smoking habit was also found to be causally related to serum MCP-3 level(β = 0.292; SE = 0.093; p = 0.002). All of the causality mentioned above remained stable in sensitivity tests. Based on the analysis results and fundamental medicine theories around macrophage-induced inflammation in degenerative intervertebral discs, we have constructed a new mechanism that long-term smoking could induce an increase in serum MCP-3 level, promoting the gathering and activation of monocyte macrophages. Furthermore, the recruited macrophages led to an increase in local IL-1β within the intervertebral disc, ultimately exacerbating the process of intervertebral disc degeneration. What we have found is expected to accelerate the development of prevention and treatment of intervertebral disc degeneration.
Collapse
Affiliation(s)
| | | | - Xiaojian Ye
- Department of Orthopedics, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| |
Collapse
|
4
|
Marvanova A, Kasik P, Elsnicova B, Tibenska V, Galatik F, Hornikova D, Zvolska V, Vebr P, Vodicka P, Hejnova L, Matous P, Szeiff Bacova B, Sykora M, Novotny J, Neuzil J, Kolar F, Novakova O, Zurmanova JM. Continuous short-term acclimation to moderate cold elicits cardioprotection in rats, and alters β-adrenergic signaling and immune status. Sci Rep 2023; 13:18287. [PMID: 37880253 PMCID: PMC10600221 DOI: 10.1038/s41598-023-44205-4] [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: 07/04/2023] [Accepted: 10/04/2023] [Indexed: 10/27/2023] Open
Abstract
Moderate cold acclimation (MCA) is a non-invasive intervention mitigating effects of various pathological conditions including myocardial infarction. We aim to determine the shortest cardioprotective regimen of MCA and the response of β1/2/3-adrenoceptors (β-AR), its downstream signaling, and inflammatory status, which play a role in cell-survival during myocardial infarction. Adult male Wistar rats were acclimated (9 °C, 1-3-10 days). Infarct size, echocardiography, western blotting, ELISA, mitochondrial respirometry, receptor binding assay, and quantitative immunofluorescence microscopy were carried out on left ventricular myocardium and brown adipose tissue (BAT). MultiPlex analysis of cytokines and chemokines in serum was accomplished. We found that short-term MCA reduced myocardial infarction, improved resistance of mitochondria to Ca2+-overload, and downregulated β1-ARs. The β2-ARs/protein kinase B/Akt were attenuated while β3-ARs translocated on the T-tubular system suggesting its activation. Protein kinase G (PKG) translocated to sarcoplasmic reticulum and phosphorylation of AMPKThr172 increased after 10 days. Principal component analysis revealed a significant shift in cytokine/chemokine serum levels on day 10 of acclimation, which corresponds to maturation of BAT. In conclusion, short-term MCA increases heart resilience to ischemia without any negative side effects such as hypertension or hypertrophy. Cold-elicited cardioprotection is accompanied by β1/2-AR desensitization, activation of the β3-AR/PKG/AMPK pathways, and an immunomodulatory effect.
Collapse
Affiliation(s)
- Aneta Marvanova
- Faculty of Science, Department of Physiology, Charles University, Vinicna 7, 128 00, Prague 2, Czech Republic
| | - Petr Kasik
- Faculty of Science, Department of Physiology, Charles University, Vinicna 7, 128 00, Prague 2, Czech Republic
| | - Barbara Elsnicova
- Faculty of Science, Department of Physiology, Charles University, Vinicna 7, 128 00, Prague 2, Czech Republic
| | - Veronika Tibenska
- Faculty of Science, Department of Physiology, Charles University, Vinicna 7, 128 00, Prague 2, Czech Republic
| | - František Galatik
- Faculty of Science, Department of Physiology, Charles University, Vinicna 7, 128 00, Prague 2, Czech Republic
| | - Daniela Hornikova
- Faculty of Science, Department of Physiology, Charles University, Vinicna 7, 128 00, Prague 2, Czech Republic
| | - Veronika Zvolska
- Faculty of Science, Department of Physiology, Charles University, Vinicna 7, 128 00, Prague 2, Czech Republic
| | - Pavel Vebr
- Faculty of Science, Department of Physiology, Charles University, Vinicna 7, 128 00, Prague 2, Czech Republic
| | - Petr Vodicka
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Libechov, Czech Republic
| | - Lucie Hejnova
- Faculty of Science, Department of Physiology, Charles University, Vinicna 7, 128 00, Prague 2, Czech Republic
| | - Petr Matous
- First Faculty of Medicine, Center for Advanced Preclinical Imaging (CAPI), Charles University, Prague, Czech Republic
| | - Barbara Szeiff Bacova
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Matus Sykora
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Jiri Novotny
- Faculty of Science, Department of Physiology, Charles University, Vinicna 7, 128 00, Prague 2, Czech Republic
| | - Jiri Neuzil
- Faculty of Science, Department of Physiology, Charles University, Vinicna 7, 128 00, Prague 2, Czech Republic
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czech Republic
- School of Pharmacy and Medical Science, Griffith University, Southport, QLD, Australia
| | - Frantisek Kolar
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Olga Novakova
- Faculty of Science, Department of Physiology, Charles University, Vinicna 7, 128 00, Prague 2, Czech Republic
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Jitka M Zurmanova
- Faculty of Science, Department of Physiology, Charles University, Vinicna 7, 128 00, Prague 2, Czech Republic.
| |
Collapse
|
5
|
Chang TT, Chen C, Chen JW. CCL7 as a novel inflammatory mediator in cardiovascular disease, diabetes mellitus, and kidney disease. Cardiovasc Diabetol 2022; 21:185. [PMID: 36109744 PMCID: PMC9479413 DOI: 10.1186/s12933-022-01626-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/09/2022] [Indexed: 11/10/2022] Open
Abstract
AbstractChemokines are key components in the pathology of chronic diseases. Chemokine CC motif ligand 7 (CCL7) is believed to be associated with cardiovascular disease, diabetes mellitus, and kidney disease. CCL7 may play a role in inflammatory events by attracting macrophages and monocytes to further amplify inflammatory processes and contribute to disease progression. However, CCL7-specific pathological signaling pathways need to be further confirmed in these chronic diseases. Given the multiple redundancy system among chemokines and their receptors, further experimental and clinical studies are needed to clarify whether direct CCL7 inhibition mechanisms could be a promising therapeutic approach to attenuating the development of cardiovascular disease, diabetes mellitus, and kidney disease.
Collapse
|
6
|
Aziz NS, Ahmad A, Yusop N. Angiogenic and Migratory Gene Expression Analysis of Stem Cells From Exfoliated Deciduous Teeth for Wound Repair Application. Curr Stem Cell Res Ther 2022; 17:466-479. [PMID: 35189800 DOI: 10.2174/1574888x17666220221142524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 11/03/2021] [Accepted: 12/21/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND The migration and differentiation of stem cells take place during the reparative phase of the healing cascade. Chemokine ligands and receptors are the key players in the homing process dur-ing the early stage of capillary morphogenesis. Stem cells from exfo-liated deciduous teeth are known to possess a huge potential benefit for tissue regeneration. However, the gene expression of SHED en-gaging in angiogenesis and migratory activity during tissue healing is not fully understood. This study aims to assess the gene expression of SHED following in-vitro angiogenesis and migratory induction protocol. METHODS Scratch test assay was conducted following an angiogenic induction of SHED by supplementation of EGM-2 and VEGF. For the detection of migratory cell markers, angiogenic markers, and stem cell markers, RNA samples were extracted on day 1, 3, 7, 10, and 14 after the angiogenic induction in a transwell chamber, followed by RT-PCR analysis. RESULTS The findings sug-gested that SHED forming endothelial cells at higher capacity under an immature state with higher seeding density. SHED undergoing angiogenesis and migratory activity showed elevated IL-8, CCR1, CXCR4 and CCL28 expression. CCR1 expression significantly in-creased in the A+M+ group (p<0.05). CONCLUSION The gene expres-sion of these chemokines, particularly CCR1, which closely represent cellular migration, suggests the potential use of SHED for cell-based therapy to enhance tissue repair.
Collapse
Affiliation(s)
- Nur Syazwani Aziz
- Postgraduate Unit, School of Dentistry, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Azlina Ahmad
- Basic Sciences and Oral Biology Unit, School of Dentistry, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Norhayati Yusop
- Basic Sciences and Oral Biology Unit, School of Dentistry, Universiti Sains Malaysia, Kelantan, Malaysia
| |
Collapse
|
7
|
Dukinfield M, Maniati E, Reynolds LE, Aubdool A, Baliga RS, D'Amico G, Maiques O, Wang J, Bedi KC, Margulies KB, Sanz‐Moreno V, Hobbs A, Hodivala‐Dilke K. Repurposing an anti-cancer agent for the treatment of hypertrophic heart disease. J Pathol 2019; 249:523-535. [PMID: 31424556 PMCID: PMC6900130 DOI: 10.1002/path.5340] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 08/14/2019] [Accepted: 08/15/2019] [Indexed: 02/06/2023]
Abstract
Coronary microvascular dysfunction combined with maladaptive cardiomyocyte morphology and energetics is a major contributor to heart failure advancement. Thus, dually enhancing cardiac angiogenesis and targeting cardiomyocyte function to slow, or reverse, the development of heart failure is a logical step towards improved therapy. We present evidence for the potential to repurpose a former anti-cancer Arg-Gly-Asp (RGD)-mimetic pentapeptide, cilengitide, here used at low doses. Cilengitide targets αvβ3 integrin and this protein is upregulated in human dilated and ischaemic cardiomyopathies. Treatment of mice after abdominal aortic constriction (AAC) surgery with low-dose cilengitide (ldCil) enhances coronary angiogenesis and directly affects cardiomyocyte hypertrophy with an associated reduction in disease severity. At a molecular level, ldCil treatment has a direct effect on cardiac endothelial cell transcriptomic profiles, with a significant enhancement of pro-angiogenic signalling pathways, corroborating the enhanced angiogenic phenotype after ldCil treatment. Moreover, ldCil treatment of Angiotensin II-stimulated AngII-stimulated cardiomyocytes significantly restores transcriptomic profiles similar to those found in normal human heart. The significance of this finding is enhanced by transcriptional similarities between AngII-treated cardiomyocytes and failing human hearts. Taken together, our data provide evidence supporting a possible new strategy for improved heart failure treatment using low-dose RGD-mimetics with relevance to human disease. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Matthew Dukinfield
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse SquareLondonUK
| | - Eleni Maniati
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse SquareLondonUK
| | - Louise E Reynolds
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse SquareLondonUK
| | - Aisah Aubdool
- William Harvey Research Institute, Queen Mary University of London, Charterhouse SquareLondonUK
| | - Reshma S Baliga
- William Harvey Research Institute, Queen Mary University of London, Charterhouse SquareLondonUK
| | - Gabriela D'Amico
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse SquareLondonUK
| | - Oscar Maiques
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse SquareLondonUK
| | - Jun Wang
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse SquareLondonUK
| | - Kenneth C Bedi
- Perelman School of MedicineUniversity of Pennsylvania, Translational Research CenterPhiladelphiaPAUSA
| | - Kenneth B Margulies
- Perelman School of MedicineUniversity of Pennsylvania, Translational Research CenterPhiladelphiaPAUSA
| | - Victoria Sanz‐Moreno
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse SquareLondonUK
| | - Adrian Hobbs
- William Harvey Research Institute, Queen Mary University of London, Charterhouse SquareLondonUK
| | - Kairbaan Hodivala‐Dilke
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse SquareLondonUK
| |
Collapse
|
8
|
Effect of Pregnancy and Delivery on Cytokine Expression in a Mouse Model of Pelvic Organ Prolapse. Female Pelvic Med Reconstr Surg 2018; 23:449-456. [PMID: 28248847 DOI: 10.1097/spv.0000000000000394] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECTIVES The aim of this study was to determine the effect of pregnancy and delivery mode on cytokine expression in the pelvic organs and serum of lysyl oxidase like-1 knockout (LOXL1 KO) mice, which develop pelvic organ prolapse after delivery. METHODS Bladder, urethra, vagina, rectum, and blood were harvested from female LOXL1 KO mice during pregnancy, after vaginal or cesarean delivery, and from sham cesarean and unmanipulated controls. Pelvic organs and blood were also harvested from pregnant and vaginally delivered wild-type (WT) mice and from unmanipulated female virgin WT controls. Specimens were assessed using quantitative real-time reverse transcription polymerase chain reaction and/or enzyme-linked immunosorbent assay. RESULTS Both CXCL12 and CCL7 mRNA were significantly up-regulated in the vagina, urethra, bladder, and rectum of pregnant LOXL1 KO mice compared with pregnant WT mice, suggesting systemic dysregulation of both of these cytokines in LOXL1 KO mice as a response to pregnancy.The differences in cytokine expression between LOXL1 KO and WT mice in pregnancy persisted after vaginal delivery. CCL7 gene expression increases faster and to a greater extent in LOXL1 KO mice, translating to longer lasting increases in CCL7 in serum of LOXL1 KO mice after vaginal delivery, compared with pregnant mice. CONCLUSIONS Lysyl oxidase like-1 KO mice have an increased cytokine response to pregnancy perhaps because they are less able to reform and re-cross-link stretched elastin to accommodate pups, and this resultant tissue stretches during pregnancy. The up-regulation of CCL7 after delivery could provide an indicator of level of childbirth injury, to which the urethra and vagina seem to be particularly vulnerable.
Collapse
|
9
|
Yanashima K, Chieosilapatham P, Yoshimoto E, Okumura K, Ogawa H, Niyonsaba F. Innate defense regulator IDR-1018 activates human mast cells through G protein-, phospholipase C-, MAPK- and NF-ĸB-sensitive pathways. Immunol Res 2018; 65:920-931. [PMID: 28653285 DOI: 10.1007/s12026-017-8932-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Host defense (antimicrobial) peptides not only display antimicrobial activities against numerous pathogens but also exert a broader spectrum of immune-modulating functions. Innate defense regulators (IDRs) are a class of host defense peptides synthetically developed from natural or endogenous cationic host defense peptides. Of the IDRs developed to date, IDR-1018 is more efficient not only in killing bacteria but also in regulating the various functions of macrophages and neutrophils and accelerating the wound healing process. Because mast cells intimately participate in wound healing and a number of host defense peptides involved in wound healing are also known to activate mast cells, this study aimed to investigate the effects of IDR-1018 on mast cell activation. Here, we showed that IDR-1018 induced the degranulation of LAD2 human mast cells and caused their production of leukotrienes, prostaglandins and various cytokines and chemokines, including granulocyte-macrophage colony-stimulating factor, interleukin-8, monocyte chemoattractant protein-1 and -3, macrophage-inflammatory protein-1α and -1β, and tumor necrosis factor-α. Furthermore, IDR-1018 increased intracellular calcium mobilization and induced mast cell chemotaxis. The mast cell activation was markedly suppressed by pertussis toxin, U-73122, U0126, SB203580, JNK inhibitor II, and NF-κB activation inhibitor II, suggesting the involvement of G-protein, phospholipase C, ERK, p38, JNK and NF-κB pathways, respectively, in IDR-1018-induced mast cell activation. Notably, we confirmed that IDR-1018 caused the phosphorylation of MAPKs and IκB. Altogether, the current study suggests a novel immunomodulatory role of IDR-1018 through its ability to recruit and activate human mast cells at the sites of inflammation and wounds. HIGHLIGHTS We report that IDR-1018 stimulates various functions of human mast cells. IDR-1018-induced mast cell activation is mediated through G protein, PLC, MAPK and NF-κB pathways. IDR-1018 will be a useful therapeutic agent for wound healing.
Collapse
Affiliation(s)
- Kensuke Yanashima
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Panjit Chieosilapatham
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.,Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Eri Yoshimoto
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Ko Okumura
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Hideoki Ogawa
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - François Niyonsaba
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan. .,Faculty of International Liberal Arts, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
| |
Collapse
|
10
|
Hinderer S, Sudrow K, Schneider M, Holeiter M, Layland SL, Seifert M, Schenke-Layland K. Surface functionalization of electrospun scaffolds using recombinant human decorin attracts circulating endothelial progenitor cells. Sci Rep 2018; 8:110. [PMID: 29311692 PMCID: PMC5758628 DOI: 10.1038/s41598-017-18382-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 12/11/2017] [Indexed: 01/25/2023] Open
Abstract
Decorin (DCN) is an important small leucine-rich proteoglycan present in the extracellular matrix (ECM) of many organs and tissues. Endothelial progenitor cells (EPCs) are able to interact with the surrounding ECM and bind to molecules such as DCN. Here, we recombinantly produced full-length human DCN under good laboratory practice (GLP) conditions, and after detailed immunological characterization, we investigated its potential to attract murine and human EPCs (mEPCs and hECFCs). Electrospun polymeric scaffolds were coated with DCN or stromal cell-derived factor-1 (SDF-1α) and were then dynamically cultured with both cell types. Cell viability was assessed via imaging flow cytometry. The number of captured cells was counted and compared with the non-coated controls. To characterize cell-scaffold interactions, immunofluorescence staining and scanning electron microscopy analyses were performed. We identified that DCN reduced T cell responses and attracted innate immune cells, which are responsible for ECM remodeling. A significantly higher number of EPCs attached on DCN- and SDF-1α-coated scaffolds, when compared with the uncoated controls. Interestingly, DCN showed a higher attractant effect on hECFCs than SDF-1α. Here, we successfully demonstrated DCN as promising EPC-attracting coating, which is particularily interesting when aiming to generate off-the-shelf biomaterials with the potential of in vivo cell seeding.
Collapse
Affiliation(s)
- Svenja Hinderer
- Department of Cell and Tissue Engineering, Fraunhofer-Institute for Interfacial Engineering and Biotechnology (IGB), 70569, Stuttgart, Germany
- Department of Women´s Health, Research Institute for Women's Health, Eberhard-Karls-University Tübingen, 72076, Tübingen, Germany
| | - Katrin Sudrow
- Institute of Medical Immunology and Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353, Berlin, Germany
| | - Maria Schneider
- Institute of Medical Immunology and Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353, Berlin, Germany
| | - Monika Holeiter
- Department of Women´s Health, Research Institute for Women's Health, Eberhard-Karls-University Tübingen, 72076, Tübingen, Germany
| | - Shannon Lee Layland
- Department of Cell and Tissue Engineering, Fraunhofer-Institute for Interfacial Engineering and Biotechnology (IGB), 70569, Stuttgart, Germany
- Department of Women´s Health, Research Institute for Women's Health, Eberhard-Karls-University Tübingen, 72076, Tübingen, Germany
| | - Martina Seifert
- Institute of Medical Immunology and Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353, Berlin, Germany
| | - Katja Schenke-Layland
- Department of Cell and Tissue Engineering, Fraunhofer-Institute for Interfacial Engineering and Biotechnology (IGB), 70569, Stuttgart, Germany.
- Department of Women´s Health, Research Institute for Women's Health, Eberhard-Karls-University Tübingen, 72076, Tübingen, Germany.
- Department of Medicine/Cardiology, Cardiovascular Research Laboratories, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
| |
Collapse
|
11
|
Li X, He X, Yin Y, Wu R, Tian B, Chen F. Administration of signalling molecules dictates stem cell homing for in situ regeneration. J Cell Mol Med 2017; 21:3162-3177. [PMID: 28767189 PMCID: PMC5706509 DOI: 10.1111/jcmm.13286] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Accepted: 05/29/2017] [Indexed: 12/13/2022] Open
Abstract
Ex vivo-expanded stem cells have long been a cornerstone of biotherapeutics and have attracted increasing attention for treating intractable diseases and improving tissue regeneration. However, using exogenous cellular materials to develop restorative treatments for large numbers of patients has become a major concern for both economic and safety reasons. Advances in cell biological research over the past two decades have expanded the potential for using endogenous stem cells during wound healing processes, and in particular, recent insight into stem cell movement and homing has prompted regenerative research and therapy based on recruiting endogenous cells. Inspired by the natural healing process, artificial administration of specific chemokines as signals systemically or at the injury site, typically using biomaterials as vehicles, is a state-of-the-art strategy that potentiates stem cell homing and recreates an anti-inflammatory and immunomodulatory microenvironment to enhance in situ tissue regeneration. However, pharmacologically coaxing endogenous stem cells to act as therapeutics in the field of biomedicine remains in the early stages; its efficacy is limited by the lack of innovative methodologies for chemokine presentation and release. This review describes how to direct the homing of endogenous stem cells via the administration of specific signals, with a particular emphasis on targeted signalling molecules that regulate this homing process, to enhance in situ tissue regeneration. We also provide an outlook on and critical considerations for future investigations to enhance stem cell recruitment and harness the reparative potential of these recruited cells as a clinically relevant cell therapy.
Collapse
Affiliation(s)
- Xuan Li
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral DiseasesDepartment of PeriodontologySchool of StomatologyFourth Military Medical UniversityXi'anChina
| | - Xiao‐Tao He
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral DiseasesDepartment of PeriodontologySchool of StomatologyFourth Military Medical UniversityXi'anChina
| | - Yuan Yin
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral DiseasesDepartment of PeriodontologySchool of StomatologyFourth Military Medical UniversityXi'anChina
| | - Rui‐Xin Wu
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral DiseasesDepartment of PeriodontologySchool of StomatologyFourth Military Medical UniversityXi'anChina
| | - Bei‐Min Tian
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral DiseasesDepartment of PeriodontologySchool of StomatologyFourth Military Medical UniversityXi'anChina
| | - Fa‐Ming Chen
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral DiseasesDepartment of PeriodontologySchool of StomatologyFourth Military Medical UniversityXi'anChina
| |
Collapse
|
12
|
Du W, Li X, Chi Y, Ma F, Li Z, Yang S, Song B, Cui J, Ma T, Li J, Tian J, Yang Z, Feng X, Chen F, Lu S, Liang L, Han ZB, Han ZC. VCAM-1+ placenta chorionic villi-derived mesenchymal stem cells display potent pro-angiogenic activity. Stem Cell Res Ther 2016; 7:49. [PMID: 27044487 PMCID: PMC4820943 DOI: 10.1186/s13287-016-0297-0] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 01/16/2016] [Accepted: 02/22/2016] [Indexed: 01/25/2023] Open
Abstract
Introduction Mesenchymal stem cells (MSCs) represent a heterogeneous cell population that is promising for regenerative medicine. The present study was designed to assess whether VCAM-1 can be used as a marker of MSC subpopulation with superior angiogenic potential. Methods MSCs were isolated from placenta chorionic villi (CV). The VCAM-1+/− CV-MSCs population were separated by Flow Cytometry and subjected to a comparative analysis for their angiogenic properties including angiogenic genes expression, vasculo-angiogenic abilities on Matrigel in vitro and in vivo, angiogenic paracrine activities, cytokine array, and therapeutic angiogenesis in vascular ischemic diseases. Results Angiogenic genes, including HGF, ANG, IL8, IL6, VEGF-A, TGFβ, MMP2 and bFGF, were up-regulated in VCAM-1+CV-MSCs. Consistently, angiogenic cytokines especially HGF, IL8, angiogenin, angiopoitin-2, μPAR, CXCL1, IL-1β, IL-1α, CSF2, CSF3, MCP-3, CTACK, and OPG were found to be significantly increased in VCAM-1+ CV-MSCs. Moreover, VCAM-1+CV-MSCs showed remarkable vasculo-angiogenic abilities by angiogenesis analysis with Matrigel in vitro and in vivo and the conditioned medium of VCAM-1+ CV-MSCs exerted markedly pro-proliferative and pro-migratory effects on endothelial cells compared to VCAM-1−CV-MSCs. Finally, transplantation of VCAM-1+CV-MSCs into the ischemic hind limb of BALB/c nude mice resulted in a significantly functional improvement in comparison with VCAM-1−CV-MSCs transplantation. Conclusions VCAM-1+CV-MSCs possessed a favorable angiogenic paracrine activity and displayed therapeutic efficacy on hindlimb ischemia. Our results suggested that VCAM-1+CV-MSCs may represent an important subpopulation of MSC for efficient therapeutic angiogenesis. Electronic supplementary material The online version of this article (doi:10.1186/s13287-016-0297-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wenjing Du
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, No.288, Nanjing Road, Heping District, Tianjin, 300020, China
| | - Xue Li
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, No.288, Nanjing Road, Heping District, Tianjin, 300020, China
| | - Ying Chi
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, No.288, Nanjing Road, Heping District, Tianjin, 300020, China
| | - Fengxia Ma
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, No.288, Nanjing Road, Heping District, Tianjin, 300020, China
| | - Zongjin Li
- Beijing Institute of Health and Stem Cells, No.1 Kangding Road, BDA, Beijing, 100176, China
| | - Shaoguang Yang
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, No.288, Nanjing Road, Heping District, Tianjin, 300020, China
| | - Baoquan Song
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, No.288, Nanjing Road, Heping District, Tianjin, 300020, China
| | - Junjie Cui
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, No.288, Nanjing Road, Heping District, Tianjin, 300020, China
| | - Tao Ma
- National Engineering Research Center of Cell Products, No.80, Fourth Avenue, TEDA, Tianjin, 300457, China
| | - Juanjuan Li
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, No.288, Nanjing Road, Heping District, Tianjin, 300020, China
| | - Jianjian Tian
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, No.288, Nanjing Road, Heping District, Tianjin, 300020, China
| | - Zhouxin Yang
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, No.288, Nanjing Road, Heping District, Tianjin, 300020, China
| | - Xiaoming Feng
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, No.288, Nanjing Road, Heping District, Tianjin, 300020, China
| | - Fang Chen
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, No.288, Nanjing Road, Heping District, Tianjin, 300020, China
| | - Shihong Lu
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, No.288, Nanjing Road, Heping District, Tianjin, 300020, China
| | - Lu Liang
- Beijing Institute of Health and Stem Cells, No.1 Kangding Road, BDA, Beijing, 100176, China
| | - Zhi-Bo Han
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, No.288, Nanjing Road, Heping District, Tianjin, 300020, China.
| | - Zhong-Chao Han
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, No.288, Nanjing Road, Heping District, Tianjin, 300020, China. .,Beijing Institute of Health and Stem Cells, No.1 Kangding Road, BDA, Beijing, 100176, China. .,National Engineering Research Center of Cell Products, No.80, Fourth Avenue, TEDA, Tianjin, 300457, China.
| |
Collapse
|
13
|
An antimicrobial peptide with angiogenic properties, AG-30/5C, activates human mast cells through the MAPK and NF-κB pathways. Immunol Res 2015; 64:594-603. [DOI: 10.1007/s12026-015-8759-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
14
|
Tissue factor pathway inhibitor gene transfer prevents vascular smooth muscle cell proliferation by interfering with the MCP-3/CCR2 pathway. J Transl Med 2015; 95:1246-57. [PMID: 26302185 DOI: 10.1038/labinvest.2015.106] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 06/12/2015] [Accepted: 06/16/2015] [Indexed: 01/13/2023] Open
Abstract
Increased vascular smooth muscle cell (VSMC) proliferation substantially contributes to the pathogenesis of atherosclerosis and intimal hyperplasia after vascular injury. The importance of inflammation in VSMC proliferation is now being recognized. Preventing the inflammatory response is one therapeutic strategy that can be used to inhibit atherosclerosis in the clinic. The present study, using RNA interference and gene transfer techniques, was conducted to investigate the effect of monocyte chemotactic protein-3 (MCP-3) on VSMC proliferation that is a result of TNF-α stimulation, and whether overexpression of the tissue factor pathway inhibitor (TFPI) gene could prevent VSMC proliferation by blocking the MCP-3/CC chemokine receptor 2 (CCR2) pathway. Mouse VSMCs were infected in vitro with recombinant adenoviruses containing either mouse MCP-3-shRNA (Ad-MCP-3-shRNA), the TFPI gene (Ad-TFPI), or the negative control, which was shRNA encoding the sequence for EGFP (Ad-EGFP) or DMEM only. The cells were then stimulated with TNF-α for different time periods on the third day after gene transfer. The data show that VSMC proliferation in the Ad-MCP-3-shRNA and Ad-TFPI groups was markedly decreased using BrdU ELISA and MTT assays; MCP-3-shRNA and TFPI inhibited the expression of MCP-3 and CCR2 after long-term stimulation and inhibited the phosphorylation of ERK1/2 and AKT after short-term stimulation, as shown by ELISA and western blot analysis. This study provides convincing evidence that clarifies the effect of the proinflammatory factor MCP-3 in promoting VSMC proliferation. Our data also show, for the first time, that TFPI has an anti-proliferative role in TNF-α stimulated-VSMCs at least partly by interfering with the MCP-3/CCR2 pathway and then via suppression of the ERK1/2 and PI3K/AKT signaling pathways. We conclude that TFPI gene transfer may be a safe and effective therapeutic tool for treating atherosclerosis and intimal hyperplasia.
Collapse
|
15
|
Nagineni CN, Kommineni VK, Ganjbaksh N, Nagineni KK, Hooks JJ, Detrick B. Inflammatory Cytokines Induce Expression of Chemokines by Human Retinal Cells: Role in Chemokine Receptor Mediated Age-related Macular Degeneration. Aging Dis 2015; 6:444-55. [PMID: 26618046 PMCID: PMC4657816 DOI: 10.14336/ad.2015.0323] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 03/23/2015] [Indexed: 12/18/2022] Open
Abstract
Chemokine reeptor-3 (CCR-3) was shown to be associated with choroidal neovascularization (CNV) in age-related macular degeneration (AMD). AMD is a vision threatening retinal disease that affects the aging population world-wide. Retinal pigment epithelium and choroid in the posterior part of the retina are the key tissues targeted in the pathogenesis of CNV in AMD. We used human retinal pigment epithelial (HRPE) and choroidal fibroblast (HCHF) cells, prepared from aged adult human donor eyes, to evaluate the expression of major CCR-3 ligands, CCL-5, CCL -7, CCL-11,CCL-24 and CCL-26. Microarray analysis of gene expression in HRPE cells treated with inflammatory cytokine mix (ICM= IFN-γ+TNF-α+IL-1β) revealed 75 and 23-fold increase in CCL-5 and CCL-7 respectively, but not CCL-11, CCL-24 and CCL-26. Chemokine secretion studies of the production of CCL5 and CCL7 by HRPE corroborated with the gene expression analysis data. When the HRPE cells were treated with either individual cytokines or the ICM, both CCL-5 and CCL-7 were produced in a dose dependent manner. Similar to the gene expression data, the ICM did not enhance HRPE production of CCL-11, CCL-24 and CCL-26. CCL-11 and CCL-26 were increased with IL-4 treatment and this HRPE production was augmented in the presence of TNF-α and IL1β. When HCHF cells were treated with either individual cytokines or the ICM, both CCL-5 and CCL-7 were produced in a dose dependent fashion. IL-4 induced low levels of CCL-11 and CCL-26 in HCHF and this production was significantly enhanced by TNF-α. Under these conditions, neither HRPE nor HCHF were demonstrated to produce CCL-24. These data demonstrate that chronic inflammation triggers CCL-5 and CCL-7 release by HRPE and HCHF and the subsequent interactions with CCR3 may participate in pathologic processes in AMD.
Collapse
Affiliation(s)
| | - Vijay K. Kommineni
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nader Ganjbaksh
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - John J. Hooks
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Barbara Detrick
- Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
16
|
Ledford KJ, Murphy N, Zeigler F, Bartel RL, Tubo R. Therapeutic potential of ixmyelocel-T, an expanded autologous multicellular therapy for treatment of ischemic cardiovascular diseases. Stem Cell Res Ther 2015; 6:25. [PMID: 25889271 PMCID: PMC4413547 DOI: 10.1186/s13287-015-0007-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 02/09/2015] [Accepted: 02/09/2015] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION Bone marrow derived cellular therapies are an emerging approach to promoting therapeutic angiogenesis in ischemic cardiovascular disease. However, the percentage of regenerative cells in bone marrow mononuclear cells (BMMNCs) is small, and large amounts of BMMNCs are required. Ixmyelocel-T, an expanded autologous multicellular therapy, is manufactured from a small sample of bone marrow aspirate. Ixmyelocel-T contains expanded populations of mesenchymal stromal cells (MSCs) and M2-like macrophages, as well as many of the CD45+ cells found in the bone marrow. It is hypothesized that this expanded multi-cellular therapy would induce angiogenesis and endothelial repair. METHODS A rat model of hind limb ischemia was used to determine the effects of ixmyelocel-T on blood flow recovery. To further determine the effects on endothelial cells, ixmyelocel-T was co-cultured with human umbilical vein endothelial cells (HUVEC) in non-contacting Transwell® inserts. RESULTS Co-culture of HUVECs with ixmyelocel-T resulted secretion of a variety of pro-angiogenic factors. HUVECs stimulated by ixmyelocel-T exhibited enhanced migration, proliferation, and branch formation. Ixmyelocel-T co-culture also resulted in increased endothelial nitric oxide synthase (eNOS) expression and nitric oxide (NO) production. In tumor necrosis factor alpha (TNFα)-stimulated HUVECs, ixmyelocel-T co-culture decreased apoptosis and reactive oxygen species generation, increased super oxide dismutase activity, and decreased nuclear factor kappa B (NFκB) activation. Treatment with ixmyelocel-T in a rat model of hind limb ischemia resulted in significantly increased blood flow perfusion and capillary density, gene expression and plasma levels of the anti-inflammatory cytokine interleukin (IL)-10, plasma nitrates, plasma platelet-derived growth factor (PDGF)-BB, vascular endothelial growth factor (VEGF) expression, and significantly decreased plasma thiobarbituric acid reactive substances (TBARS). CONCLUSIONS This work demonstrates that ixmyelocel-T interacts with endothelial cells in a paracrine manner, resulting in angiogenesis and endothelial protection. This data suggests that ixmyelocel-T could be useful for promoting of angiogenesis and tissue repair in ischemic cardiovascular diseases. In conclusion, ixmyelocel-T therapy may provide a new aspect of therapeutic angiogenesis in this patient population where expanded populations of regenerative cells might be required.
Collapse
Affiliation(s)
- Kelly J Ledford
- Aastrom Biosciences, Domino's Farms, Lobby K 24 Frank Lloyd Wright Drive, Ann Arbor, MI, 48105, USA.
| | - Nikki Murphy
- Aastrom Biosciences, Domino's Farms, Lobby K 24 Frank Lloyd Wright Drive, Ann Arbor, MI, 48105, USA.
| | - Frank Zeigler
- Aastrom Biosciences, Domino's Farms, Lobby K 24 Frank Lloyd Wright Drive, Ann Arbor, MI, 48105, USA.
| | - Ronnda L Bartel
- Aastrom Biosciences, Domino's Farms, Lobby K 24 Frank Lloyd Wright Drive, Ann Arbor, MI, 48105, USA.
| | - Ross Tubo
- Aastrom Biosciences, Domino's Farms, Lobby K 24 Frank Lloyd Wright Drive, Ann Arbor, MI, 48105, USA.
| |
Collapse
|
17
|
Liu J, Li W, Wang Y, Fan W, Li P, Lin W, Yang D, Fang R, Feng M, Hu C, Du Z, Wu G, Xiang AP. Islet-1 overexpression in human mesenchymal stem cells promotes vascularization through monocyte chemoattractant protein-3. Stem Cells 2015; 32:1843-54. [PMID: 24578274 DOI: 10.1002/stem.1682] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 01/27/2014] [Accepted: 02/11/2014] [Indexed: 01/14/2023]
Abstract
The LIM-homeobox transcription factor islet-1 (ISL1) has been proposed to mark a cardiovascular progenitor cell lineage that gives rise to cardiomyocytes, endothelial cells, and smooth muscle cells. The aim of this study was to investigate whether forced expression of ISL1 in human mesenchymal stem cells (hMSCs) influenced the differentiation capacity and angiogenic properties of hMSCs. The lentiviral vector, EF1α-ISL1, was constructed using the Multisite Gateway System and used to transduce hMSCs. We found that ISL1 overexpression did not alter the proliferation, migration, or survival of hMSCs or affect their ability to differentiate into osteoblasts, adipocytes, cardiomyocytes, or endotheliocytes. However, ISL1-hMSCs differentiated into smooth muscle cells more efficiently than control hMSCs. Furthermore, conditioned medium from ISL1-hMSCs greatly enhanced the survival, migration, and tube-formation ability of human umbilical vein endothelial cells (HUVECs) in vitro. In vivo angiogenesis assays also showed much more vascular-like structures in the group cotransplanted with ISL1-hMSCs and HUVECs than in the group cotransplanted with control hMSCs and HUVECs. Quantitative RT-PCR and antibody arrays detected monocyte chemoattractant protein-3 (MCP3) at a higher level in conditioned medium from ISL1-hMSCs cultures than in conditioned medium from control hMSCs. Neutralization assays showed that addition of an anti-MCP3 antibody to ISL1-hMSCs-conditioned medium efficiently abolished the angiogenesis-promoting effect of ISL1-hMSCs. Our data suggest that overexpression of ISL1 in hMSCs promotes angiogenesis in vitro and in vivo through increasing secretion of paracrine factors, smooth muscle differentiation ability, and enhancing the survival of HUVECs.
Collapse
Affiliation(s)
- Jia Liu
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China; Heart Center, The Affiliated Futian Hospital of Guangdong Medical College, Shenzhen, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Cheng JW, Sadeghi Z, Levine AD, Penn MS, von Recum HA, Caplan AI, Hijaz A. The role of CXCL12 and CCL7 chemokines in immune regulation, embryonic development, and tissue regeneration. Cytokine 2014; 69:277-83. [PMID: 25034237 DOI: 10.1016/j.cyto.2014.06.007] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Revised: 05/23/2014] [Accepted: 06/04/2014] [Indexed: 12/20/2022]
Abstract
Chemotactic factors direct the migration of immune cells, multipotent stem cells, and progenitor cells under physiologic and pathologic conditions. Chemokine ligand 12 and chemokine ligand 7 have been identified and investigated in multiple studies for their role in cellular trafficking in the setting of tissue regeneration. Recent early phase clinical trials have suggested that these molecules may lead to clinical benefit in patients with chronic disease. Importantly, these two proteins may play additional significant roles in directing the migration of multipotent cells, such as mesenchymal stem cells and hematopoietic progenitor cells. This article reviews the functions of these two chemokines, focusing on recruitment to sites of injury, immune function modulation, and contributions to embryonic development. Additional research would provide valuable insight into the potential clinical application of these two proteins in stem cell therapy.
Collapse
Affiliation(s)
- Julie W Cheng
- Urology Institute, University Hospitals Case Medical Center, Department of Urology, Case Western Reserve University School of Medicine, 11100 Euclid Avenue, Cleveland, OH 44106, United States
| | - Zhina Sadeghi
- Urology Institute, University Hospitals Case Medical Center, Department of Urology, Case Western Reserve University School of Medicine, 11100 Euclid Avenue, Cleveland, OH 44106, United States
| | - Alan D Levine
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States; Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States; Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States
| | - Marc S Penn
- Skirball Laboratory for Cardiovascular Cellular Therapeutics, Summa Cardiovascular Institute, Summa Health System, 525 East Market Street, Akron, OH 44304, United States
| | - Horst A von Recum
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, United States
| | - Arnold I Caplan
- Skeletal Research Center, Department of Biology, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, United States
| | - Adonis Hijaz
- Urology Institute, University Hospitals Case Medical Center, Department of Urology, Case Western Reserve University School of Medicine, 11100 Euclid Avenue, Cleveland, OH 44106, United States.
| |
Collapse
|
19
|
Abstract
RATIONALE Long noncoding RNAs (lncRNAs) constitute a novel class of noncoding RNAs that regulate gene expression. Although recent data suggest that lncRNAs may be associated with cardiac disease, little is known about lncRNAs in the setting of myocardial ischemia. OBJECTIVE To measure lncRNAs in patients with myocardial infarction (MI). METHODS AND RESULTS We enrolled 414 patients with acute MI treated by primary percutaneous coronary intervention. Blood samples were harvested at the time of reperfusion. Expression levels of 5 lncRNAs were measured in peripheral blood cells by quantitative polymerase chain reaction: hypoxia inducible factor 1A antisense RNA 2, cyclin-dependent kinase inhibitor 2B antisense RNA 1 (ANRIL), potassium voltage-gated channel, KQT-like subfamily, member 1 opposite strand/antisense transcript 1 (KCNQ1OT1), myocardial infarction-associated transcript, and metastasis-associated lung adenocarcinoma transcript 1. Levels of hypoxia inducible factor 1A antisense RNA 2, KCNQ1OT1, and metastasis-associated lung adenocarcinoma transcript 1 were higher in patients with MI than in healthy volunteers (P<0.01), and levels of ANRIL were lower in patients with MI (P=0.003). Patients with ST-segment-elevation MI had lower levels of ANRIL (P<0.001), KCNQ1OT1 (P<0.001), myocardial infarction-associated transcript (P<0.001), and metastasis-associated lung adenocarcinoma transcript 1 (P=0.005) when compared with patients with non-ST-segment-elevation MI. Levels of ANRIL were associated with age, diabetes mellitus, and hypertension. Patients presenting within 3 hours of chest pain onset had elevated levels of hypoxia inducible factor 1A antisense RNA 2 when compared with patients presenting later on. ANRIL, KCNQ1OT1, myocardial infarction-associated transcript, and metastasis-associated lung adenocarcinoma transcript 1 were significant univariable predictors of left ventricular dysfunction as assessed by an ejection fraction ≤40% at 4-month follow-up. In multivariable and reclassification analyses, ANRIL and KCNQ1OT1 improved the prediction of left ventricular dysfunction by a model, including demographic features, clinical parameters, and cardiac biomarkers. CONCLUSIONS Levels of lncRNAs in blood cells are regulated after MI and may help in prediction of outcome. This motivates further investigation of the role of lncRNAs after MI.
Collapse
Affiliation(s)
- Mélanie Vausort
- From the Laboratory of Cardiovascular Research, Centre de Recherche Public de la Santé, Luxembourg, Luxembourg (M.V., Y.D.); and Division of Cardiology, Centre Hospitalier, Luxembourg, Luxembourg (D.R.W.)
| | - Daniel R Wagner
- From the Laboratory of Cardiovascular Research, Centre de Recherche Public de la Santé, Luxembourg, Luxembourg (M.V., Y.D.); and Division of Cardiology, Centre Hospitalier, Luxembourg, Luxembourg (D.R.W.)
| | - Yvan Devaux
- From the Laboratory of Cardiovascular Research, Centre de Recherche Public de la Santé, Luxembourg, Luxembourg (M.V., Y.D.); and Division of Cardiology, Centre Hospitalier, Luxembourg, Luxembourg (D.R.W.).
| |
Collapse
|
20
|
Jawad S, Liu B, Li Z, Katamay R, Campos M, Wei L, Sen HN, Ling D, Martinez Estrada F, Amaral J, Chan CC, Fariss R, Gordon S, Nussenblatt RB. The role of macrophage class a scavenger receptors in a laser-induced murine choroidal neovascularization model. Invest Ophthalmol Vis Sci 2013; 54:5959-70. [PMID: 23927892 DOI: 10.1167/iovs.12-11380] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Laser-induced choroidal neovascularization (CNV) is a widely used model to mimic many features of CNV resulting from wet AMD. Macrophages have been implicated in the pathogenesis of AMD. Class A scavenger receptors, scavenger receptor-A (SR-A) and macrophage receptor with collagenous domain (MARCO), are expressed on macrophages and are associated with macrophage function. The goal of this study is to examine the role of macrophage scavenger receptors in immune cell recruitment and the formation of CNV. METHODS Laser photocoagulation was performed in wild-type and knockout mice with deletion of SR-A (SR-A(-/-)), MARCO (MARCO(-/-)), or both SR-A and MARCO double knockout (DKO). Immune cell recruitment at different time points and CNV lesions at 14 days after laser treatment were evaluated through immunostaining and confocal microscopy. Microarray analysis was performed in eyes 1 day after laser injury. RESULTS Wild-type eyes showed higher chemokine/receptor expression compared with knockout eyes after laser injury. Scavenger receptor deficiency markedly impaired the recruitment of neutrophils and macrophages to CNV lesions at 1- and 3-days post laser injury, respectively. Significantly reduced CNV volumes were found in the eyes from scavenger receptor knockout mice compared with wild-type mice. CONCLUSIONS The deficiency of scavenger receptors impairs the formation of CNV and immune cell recruitment. Our findings suggest a potential role for scavenger receptors in contributing to CNV formation and inflammation in AMD.
Collapse
Affiliation(s)
- Shayma Jawad
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Yellowley C. CXCL12/CXCR4 signaling and other recruitment and homing pathways in fracture repair. BONEKEY REPORTS 2013; 2:300. [PMID: 24422056 DOI: 10.1038/bonekey.2013.34] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Accepted: 02/08/2013] [Indexed: 02/06/2023]
Abstract
Cell recruitment, migration and homing to the fracture site are essential for the inflammatory process, neovascularization, chondrogenesis, osteogenesis and ultimately bone remodeling. Mesenchymal stem cells (MSCs) are required to navigate from local sources such as the periosteum and local bone marrow, and may also be recruited from the circulation and distant bone marrow. While the local recruitment process may involve matrix binding and degradation, systemic recruitment may utilize extravasation, a process used by leukocytes to exit the vasculature. CXCL12 (stromal cell-derived factor-1 (SDF-1)), a member of the CXC family of chemokines, is thought to have an important role in cell migration at the fracture site. However, there are many molecules upregulated in the hematoma and callus that have chemotactic potential not only for inflammatory cells but also for endothelial cells and MSCs. Surprisingly, there is little direct data to support their role in cell homing during bone healing. Current therapeutics for bone regeneration utilize local or systemic stem cell transplantation. More recently, a novel strategy that involves mobilization of large numbers of endogenous stem and progenitor cells from bone marrow into the circulation has been shown to have positive effects on bone healing. A more complete understanding of the molecular mechanisms underlying cell recruitment and homing subsequent to fracture will facilitate the fine-tuning of such strategies for bone.
Collapse
Affiliation(s)
- Clare Yellowley
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis , Davis, CA, USA
| |
Collapse
|
22
|
Rolland-Turner M, Goretti E, Bousquenaud M, Léonard F, Nicolas C, Zhang L, Maskali F, Marie PY, Devaux Y, Wagner D. Adenosine stimulates the migration of human endothelial progenitor cells. Role of CXCR4 and microRNA-150. PLoS One 2013; 8:e54135. [PMID: 23326587 PMCID: PMC3541240 DOI: 10.1371/journal.pone.0054135] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Accepted: 12/10/2012] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Administration of endothelial progenitor cells (EPC) represents a promising option to regenerate the heart after myocardial infarction, but is limited because of low recruitment and engraftment in the myocardium. Mobilization and migration of EPC are mainly controlled by stromal cell-derived factor 1α (SDF-1α) and its receptor CXCR4. We hypothesized that adenosine, a cardioprotective molecule, may improve the recruitment of EPC to the heart. METHODS EPC were obtained from peripheral blood mononuclear cells of healthy volunteers. Expression of chemokines and their receptors was evaluated using microarrays, quantitative PCR, and flow cytometry. A Boyden chamber assay was used to assess chemotaxis. Recruitment of EPC to the infarcted heart was evaluated in rats after permanent occlusion of the left anterior descending coronary artery. RESULTS Microarray analysis revealed that adenosine modulates the expression of several members of the chemokine family in EPC. Among these, CXCR4 was up-regulated by adenosine, and this result was confirmed by quantitative PCR (3-fold increase, P<0.001). CXCR4 expression at the cell surface was also increased. This effect involved the A(2B) receptor. Pretreatment of EPC with adenosine amplified their migration towards recombinant SDF-1α or conditioned medium from cardiac fibroblasts. Both effects were abolished by CXCR4 blocking antibodies. Adenosine also increased CXCR4 under ischemic conditions, and decreased miR-150 expression. Binding of miR-150 to the 3' untranslated region of CXCR4 was verified by luciferase assay. Addition of pre-miR-150 blunted the effect of adenosine on CXCR4. Administration of adenosine to rats after induction of myocardial infarction stimulated EPC recruitment to the heart and enhanced angiogenesis. CONCLUSION Adenosine increases the migration of EPC. The mechanism involves A(2B) receptor activation, decreased expression of miR-150 and increased expression of CXCR4. These results suggest that adenosine may be used to enhance the capacity of EPC to revascularize the ischemic heart.
Collapse
Affiliation(s)
- Magali Rolland-Turner
- Laboratory of Cardiovascular Research, Public Research Centre – Health (CRP – Santé), Luxembourg, Luxembourg
| | - Emeline Goretti
- Laboratory of Cardiovascular Research, Public Research Centre – Health (CRP – Santé), Luxembourg, Luxembourg
| | - Mélanie Bousquenaud
- Laboratory of Cardiovascular Research, Public Research Centre – Health (CRP – Santé), Luxembourg, Luxembourg
| | - Frédérique Léonard
- Laboratory of Cardiovascular Research, Public Research Centre – Health (CRP – Santé), Luxembourg, Luxembourg
| | - Christelle Nicolas
- Laboratory of Cardiovascular Research, Public Research Centre – Health (CRP – Santé), Luxembourg, Luxembourg
| | - Lu Zhang
- Laboratory of Cardiovascular Research, Public Research Centre – Health (CRP – Santé), Luxembourg, Luxembourg
| | - Fatiha Maskali
- Nancyclotep Experimental Imaging Platform, Vandoeuvre-les-Nancy, France
| | - Pierre-Yves Marie
- Nancyclotep Experimental Imaging Platform, Vandoeuvre-les-Nancy, France
| | - Yvan Devaux
- Laboratory of Cardiovascular Research, Public Research Centre – Health (CRP – Santé), Luxembourg, Luxembourg
| | - Daniel Wagner
- Laboratory of Cardiovascular Research, Public Research Centre – Health (CRP – Santé), Luxembourg, Luxembourg
- Division of Cardiology, Centre Hospitalier, Luxembourg, Luxembourg
| |
Collapse
|
23
|
Radillo O, Norcio A, Addobbati R, Zauli G. Presence of CTAK/CCL27, MCP-3/CCL7 and LIF in human colostrum and breast milk. Cytokine 2012; 61:26-8. [PMID: 23040056 DOI: 10.1016/j.cyto.2012.09.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 09/02/2012] [Indexed: 11/30/2022]
Abstract
Human colostrum and breast milk are known to contain high levels of cytokines and chemokines, which are thought to contribute to the development of the newborn. The aim of this study was to investigate the difference in the presence and levels of 21 soluble cytokines and chemokines in paired samples of human colostrum (day 2 after delivery) and breast milk (day 4-5 after delivery) by using the multiplex technology. Of the 21 cytokine investigated in 10 pairs of samples, only β-NGF was absent in both colostrum and milk, while INF-α2, SCF and TNF-β were present in colostrum but not in human milk. As a general rule, colostrum contained higher concentrations of cytokines and chemokines with respect to breast milk. The majority of cytokines, detected in colostrum alone or in colostrum and human milk (IL-1α, IL-2Rα, IL-3, IL-16, IL-18, GRO-α, HGF, IFN-α2, M-CSF, MIF, MIG, TNF-β, SDF-1α, TRAIL) have been described in previous studies, while for the first time we describe the presence of additional cytokines either in colostrum alone (SCF) or in both colostrum and breast milk (CTAK/CCL27, MCP-3/CCL7, LIF). Our data confirm and expand previous studies showing that some cytokines/chemokines, which might contribute to the development of the gastro-intestinal and nervous systems, are overexpressed in human colostrum and breast milk, and might contribute to the development of these systems.
Collapse
Affiliation(s)
- Oriano Radillo
- Institute for Maternal and Child Health-IRCCS Burlo Garofolo, Trieste, Italy.
| | | | | | | |
Collapse
|