1
|
Yang L, Miao Z, Li N, Meng L, Feng Q, Qiao D, Wang P, Wang Y, Bai Y, Li Z, Lian S. CMTM4 promotes the motility of colon cancer cells under radiation and is associated with an unfavorable neoadjuvant chemoradiotherapy response and patient survival in rectal cancer. Oncol Lett 2025; 29:138. [PMID: 39839608 PMCID: PMC11747855 DOI: 10.3892/ol.2025.14884] [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: 06/18/2024] [Accepted: 12/02/2024] [Indexed: 01/23/2025] Open
Abstract
Neoadjuvant chemoradiotherapy (nCRT) is the standard treatment for locally advanced rectal cancer (LARC). Pathological complete regression is closely linked to disease outcomes. However, biomarkers predicting nCRT response and patient survival are lacking for LARC. In the present study, the clinical characteristics and follow-up information of 228 patients with LARC were retrospectively collected. Immunohistochemistry (IHC), reverse transcription-quantitative PCR (RT-qPCR), Kaplan-Meier and multivariate analyses were used to evaluate the expression and predict the role of CKLF-like MARVEL transmembrane domain member 4 (CMTM4) in LARC. Additionally, lentiviral short hairpin (sh)RNA was used to interfere with CMTM4 expression. The phenotype of CMTM4-knockdown LoVo cells was determined by colony formation, migration and invasion assays under irradiation (IR) treatment. RNA-sequencing (RNA-seq) analysis was also used to explore the CMTM4-regulated genes in LoVo-shCMTM4 cells compared with control cells. RT-qPCR was then used to confirm the expression of these CMTM4-regulated genes. CMTM4 expression in pre-nCRT tissues indicated an unfavorable response and a short disease-free survival (DFS) with LARC. The expression of CMTM4 significantly increased following nCRT treatment. Additionally, CMTM4 knockdown increased the proliferation, migration and invasion of colon cancer cells; however, IR disrupted the cell migration and invasion induced by CMTM4 knockdown. RNA-seq analysis, the Tumor Immune Estimation Resource database and RT-qPCR indicated that CMTM4 was involved in different signaling pathways and regulated immune-related genes such as cluster of differentiation 66b, chemokine (CXC motif) ligand 8 (CXCL8) and programmed cell death 1. Furthermore, CXCL8 expression was found to be negatively associated with CMTM4 expression in patients with LARC by IHC and RT-qPCR. CXCL8 expression on invasion margin regions in post-operative tissues was also an inferior predictor of DFS in patients with LARC. In conclusion, CMTM4 may predict the nCRT response and outcomes in patients with LARC.
Collapse
Affiliation(s)
- Lujing Yang
- Department of Pathology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Zhiting Miao
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Ningning Li
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Lin Meng
- Department of Biochemistry and Molecular Biology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Qin Feng
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Dongbo Qiao
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Ping Wang
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Yue Wang
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Yanhua Bai
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Zhongwu Li
- Department of Pathology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Shenyi Lian
- Department of Pathology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| |
Collapse
|
2
|
Lertwanakarn T, Reyes A, Salazar E, Barrientos M, Sanchez EE, Suntravat M. The Effect of Purified Opharin Isolated from the Venom of King Cobra ( Ophiophagus hannah) in Modulating Macrophage Inflammatory Responses and Vascular Integrity. Toxins (Basel) 2024; 16:550. [PMID: 39728808 DOI: 10.3390/toxins16120550] [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: 09/19/2024] [Revised: 12/07/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024] Open
Abstract
King cobra (Ophiophagus hannah) venom comprises a diverse array of proteins and peptides. However, the roles and properties of these individual components are still not fully understood. Among these, Cysteine-rich secretory proteins (CRiSPs) are recognized but not fully characterized. This study investigates the biological effects of Opharin, the CRiSP from king cobra venom (KCV). The effects of Opharin on cytokine production, specifically on IL-1β, IL-6, IL-8, TNF-α, and IL-10 release, were evaluated over 24 h in monocyte-derived macrophage (MDM) cells. Notably, the levels of these inflammatory cytokines were significantly increased over 24 h, with values higher than those observed in cells treated with crude KCV at most time points. Additionally, the in vivo Miles assay in mice revealed that Opharin increased vascular permeability by 26% compared to the negative control group. These findings highlight the Opharin's role in severe inflammatory and vascular responses observed in king cobra envenomation. Still, further research is essential to elucidate the pharmacological and toxicological effects of venom components, ultimately enhancing the clinical management of envenomation.
Collapse
Affiliation(s)
- Tuchakorn Lertwanakarn
- Department of Physiology, Faculty of Veterinary Medicine, Kasetsart University, Bangkok 10310, Thailand
| | - Armando Reyes
- National Natural Toxins Research Center (NNTRC), Texas A&M University-Kingsville, Kingsville, TX 78363, USA
| | - Emelyn Salazar
- National Natural Toxins Research Center (NNTRC), Texas A&M University-Kingsville, Kingsville, TX 78363, USA
| | - Martha Barrientos
- National Natural Toxins Research Center (NNTRC), Texas A&M University-Kingsville, Kingsville, TX 78363, USA
| | - Elda E Sanchez
- National Natural Toxins Research Center (NNTRC), Texas A&M University-Kingsville, Kingsville, TX 78363, USA
- Department of Chemistry, Texas A&M University-Kingsville, MSC 161, Kingsville, TX 78363, USA
| | - Montamas Suntravat
- National Natural Toxins Research Center (NNTRC), Texas A&M University-Kingsville, Kingsville, TX 78363, USA
- Department of Chemistry, Texas A&M University-Kingsville, MSC 161, Kingsville, TX 78363, USA
| |
Collapse
|
3
|
Zhou H, Scatena M, Tu LN, Giachelli CM, Nigam V. Monocyte adhesion to and transmigration through endothelium following cardiopulmonary bypass shearing is mediated by IL-8 signaling. Front Cardiovasc Med 2024; 11:1454302. [PMID: 39723411 PMCID: PMC11668754 DOI: 10.3389/fcvm.2024.1454302] [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: 06/24/2024] [Accepted: 11/21/2024] [Indexed: 12/28/2024] Open
Abstract
Introduction The use of cardiopulmonary bypass (CPB) can induce sterile systemic inflammation that contributes to morbidity and mortality, especially in children. Patients have been found to have increased expression of cytokines and transmigration of leukocytes during and after CPB. Previous work has demonstrated that the supraphysiologic shear stresses existing during CPB are sufficient to induce proinflammatory behavior in non-adherent monocytes. The interactions between shear stimulated monocytes and vascular endothelial cells have not been well studied and have important translational implications. With these studies, we tested the hypothesis that non-physiological shear stress experienced by monocytes during CPB affects the integrity and function of the endothelial monolayer. Methods We have used an in vitro CPB model to study the interaction between THP-1 monocyte-like cells and human neonatal dermal microvascular endothelial cells (HNDMVECs). THP-1 cells were sheared in polyvinyl chloride (PVC) tubing at 2.1 Pa, twice of the physiological shear stress, for 2 h. ELISA, adhesion and transmigration assays, qPCR, and RNA silencing were used to assess the interactions between THP-1 cells and HNDMVECs were characterized after co-culture. Results We found that sheared THP-1 cells adhered to and transmigrated through the HNDMVEC monolayer more readily than static THP-1 controls. Sheared THP-1 cells disrupted the VE-cadherin and led to the reorganization of cytoskeletal F-actin of HNDMVECs. A higher level of IL-8 was detected in the sheared THP-1 and HNDMVEC co-culture medium compared to the static THP-1 and HNDMVEC medium. Further, treating HNDMVECs with IL-8 resulted in increased adherence of non-sheared THP-1 cells, and upregulation in HNDMVECs of vascular cell adhesion molecule 1 (VCAM-1) and intercellular adhesion molecule 1 (ICAM-1). Finally, inhibition of HNDMVECs CXCR2/IL-8 receptor with Reparixin and of IL-8 expression with siRNA blocked sheared THP-1 cell adhesion to the endothelial monolayer. Conclusions These results suggest that CPB-like sheared monocytes promote IL-8 production followed by increased endothelium permeability, and monocyte adhesion and transmigration. This study revealed a novel mechanism of post-CPB inflammation and will contribute to the development of targeted therapeutics to prevent and repair the damage to neonatal patients.
Collapse
Affiliation(s)
- Hao Zhou
- Department of Bioengineering, University of Washington, Seattle, WA, United States
| | - Marta Scatena
- Department of Bioengineering, University of Washington, Seattle, WA, United States
| | - Lan N. Tu
- Seattle Children’s Hospital, Seattle, WA, United States
| | - Cecilia M. Giachelli
- Department of Bioengineering, University of Washington, Seattle, WA, United States
| | - Vishal Nigam
- Seattle Children’s Hospital, Seattle, WA, United States
- Department of Pediatrics, University of Washington, Seattle, WA, United States
| |
Collapse
|
4
|
Garcia MJ, Morales MS, Yang TS, Holden J, Bossardet OL, Palmer SA, Jhala M, Priest S, Namburu N, Beatty N, D'Empaire Salomon SE, Vancel J, Wareham LK, Padovani-Claudio DA. Adverse effects of CXCR2 deficiency in mice reared under non-gnotobiotic conditions. Sci Rep 2024; 14:26159. [PMID: 39478033 PMCID: PMC11525579 DOI: 10.1038/s41598-024-75532-9] [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: 01/22/2024] [Accepted: 10/07/2024] [Indexed: 11/02/2024] Open
Abstract
The family of pro-inflammatory and pro-angiogenic chemokines including Interleukin-8 (IL-8, aka CXCL8) and its homologues (CXCL1,2,3,5,6, and 7) exhibit promiscuous binding and activation of several G-protein-coupled receptors (i.e., CXCR2, CXCR1, and the atypical chemokine receptor (ACKR1)). A high proportion of their biological activity is attributed to CXCR2 activation, thus many CXCR2 inhibitors are in clinical trials for several chronic diseases. However, CXCR2 inhibition is often only investigated acutely in these trials or in Cxcr2-/- mice grown in gnotobiotic conditions. Since humans do not live in germ-free environments, our first goal is to highlight novel retinal and systemic observations in Cxcr2-/- mice grown in non-gnotobiotic conditions that suggest potential harmful consequences of long-term CXCR2 deficiency or blockade. Beyond confirmation of circulating blood/immune cell-related phenotypes, we report novel findings in Cxcr2-/- mice including: (1) delayed dye transit to the retinal vasculature, (2) alterations in the density and distribution of retinal vessels, astrocytes and microglia, (3) decreased electroretinogram a- and b-wave amplitudes, (4) reduced visual acuity, and (5) increased polymorphonuclear cell accumulation in vascular lumina abutting venular walls in the retina and in vital non-ocular tissues (lung and liver). Furthermore, PheWAS of CXCR2 CXCR1, and ACKR1 gene variants using data from UK Biobank participants suggest clinical associations with both retinal and vascular disease phenotypes. We conclude that chronic CXCR2 deficiency in mice contributes to functional damage to the retina and that the long-term safety of CXCR1/2 inhibitors designed for chronic use in humans should be explored before clinical adoption to safeguard sight and overall vascular health.
Collapse
Affiliation(s)
- Maximilian J Garcia
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute/Vanderbilt University Medical Center, 2311 Pierce Avenue, Nashville, TN, USA
| | - Monica S Morales
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute/Vanderbilt University Medical Center, 2311 Pierce Avenue, Nashville, TN, USA
| | - Tzushan S Yang
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Joseph Holden
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute/Vanderbilt University Medical Center, 2311 Pierce Avenue, Nashville, TN, USA
| | - Olivia L Bossardet
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute/Vanderbilt University Medical Center, 2311 Pierce Avenue, Nashville, TN, USA
| | - Samuel A Palmer
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute/Vanderbilt University Medical Center, 2311 Pierce Avenue, Nashville, TN, USA
| | - Marvarakumari Jhala
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute/Vanderbilt University Medical Center, 2311 Pierce Avenue, Nashville, TN, USA
| | - Stephen Priest
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute/Vanderbilt University Medical Center, 2311 Pierce Avenue, Nashville, TN, USA
| | - Neeraj Namburu
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute/Vanderbilt University Medical Center, 2311 Pierce Avenue, Nashville, TN, USA
| | - Nolan Beatty
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute/Vanderbilt University Medical Center, 2311 Pierce Avenue, Nashville, TN, USA
| | - Sariah E D'Empaire Salomon
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute/Vanderbilt University Medical Center, 2311 Pierce Avenue, Nashville, TN, USA
| | - Jordan Vancel
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute/Vanderbilt University Medical Center, 2311 Pierce Avenue, Nashville, TN, USA
| | - Lauren K Wareham
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute/Vanderbilt University Medical Center, 2311 Pierce Avenue, Nashville, TN, USA
| | - Dolly Ann Padovani-Claudio
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute/Vanderbilt University Medical Center, 2311 Pierce Avenue, Nashville, TN, USA.
- Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
5
|
Sajib MS, Zahra FT, Lamprou M, Akwii RG, Park JH, Osorio M, Tullar P, Doci CL, Zhang C, Huveneers S, Van Buul JD, Wang MH, Markiewski MM, Srivastava SK, Zheng Y, Gutkind JS, Hu J, Bickel U, Maeda DY, Zebala JA, Lionakis MS, Trasti S, Mikelis CM. Tumor-induced endothelial RhoA activation mediates tumor cell transendothelial migration and metastasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.22.614304. [PMID: 39372784 PMCID: PMC11451620 DOI: 10.1101/2024.09.22.614304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
The endothelial barrier plays an active role in transendothelial tumor cell migration during metastasis, however, the endothelial regulatory elements of this step remain obscure. Here we show that endothelial RhoA activation is a determining factor during this process. Breast tumor cell-induced endothelial RhoA activation is the combined outcome of paracrine IL-8-dependent and cell-to-cell contact β 1 integrin-mediated mechanisms, with elements of this pathway correlating with clinical data. Endothelial-specific RhoA blockade or in vivo deficiency inhibited the transendothelial migration and metastatic potential of human breast tumor and three murine syngeneic tumor cell lines, similar to the pharmacological blockade of the downstream RhoA pathway. These findings highlight endothelial RhoA as a potent, universal target in the tumor microenvironment for anti-metastatic treatment of solid tumors.
Collapse
|
6
|
Mohsin F, Javaid S, Tariq M, Mustafa M. Molecular immunological mechanisms of impaired wound healing in diabetic foot ulcers (DFU), current therapeutic strategies and future directions. Int Immunopharmacol 2024; 139:112713. [PMID: 39047451 DOI: 10.1016/j.intimp.2024.112713] [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: 04/04/2024] [Revised: 07/02/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024]
Abstract
Diabetic foot ulcer (DFU) is a foremost cause of amputation in diabetic patients. Consequences of DFU include infections, decline in limb function, hospitalization, amputation, and in severe cases, death. Immune cells including macrophages, regulatory T cells, fibroblasts and other damage repair cells work in sync for effective healing and in establishment of a healthy skin barrier post-injury. Immune dysregulation during the healing of wounds can result in wound chronicity. Hyperglycemic conditions in diabetic patients influence the pathophysiology of wounds by disrupting the immune system as well as promoting neuropathy and ischemic conditions, making them difficult to heal. Chronic wound microenvironment is characterized by increased expression of matrix metalloproteinases, reactive oxygen species as well as pro-inflammatory cytokines, resulting in persistent inflammation and delayed healing. Novel treatment modalities including growth factor therapies, nano formulations, microRNA based treatments and skin grafting approaches have significantly augmented treatment efficiency, demonstrating creditable efficacy in clinical practices. Advancements in local treatments as well as invasive methodologies, for instance formulated wound dressings, stem cell applications and immunomodulatory therapies have been successful in targeting the complex pathophysiology of chronic wounds. This review focuses on elucidating the intricacies of emerging physical and non-physical therapeutic interventions, delving into the realm of advanced wound care and comprehensively summarizing efficacy of evidence-based therapies for DFU currently available.
Collapse
Affiliation(s)
- Fatima Mohsin
- KAM School of Life Sciences, Forman Christian College (A Chartered University), Lahore, Pakistan.
| | - Sheza Javaid
- KAM School of Life Sciences, Forman Christian College (A Chartered University), Lahore, Pakistan.
| | - Mishal Tariq
- KAM School of Life Sciences, Forman Christian College (A Chartered University), Lahore, Pakistan.
| | - Muhammad Mustafa
- KAM School of Life Sciences, Forman Christian College (A Chartered University), Lahore, Pakistan.
| |
Collapse
|
7
|
Mamun AA, Shao C, Geng P, Wang S, Xiao J. Recent advances in molecular mechanisms of skin wound healing and its treatments. Front Immunol 2024; 15:1395479. [PMID: 38835782 PMCID: PMC11148235 DOI: 10.3389/fimmu.2024.1395479] [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: 03/04/2024] [Accepted: 05/03/2024] [Indexed: 06/06/2024] Open
Abstract
The skin, being a multifaceted organ, performs a pivotal function in the complicated wound-healing procedure, which encompasses the triggering of several cellular entities and signaling cascades. Aberrations in the typical healing process of wounds may result in atypical scar development and the establishment of a persistent condition, rendering patients more vulnerable to infections. Chronic burns and wounds have a detrimental effect on the overall quality of life of patients, resulting in higher levels of physical discomfort and socio-economic complexities. The occurrence and frequency of prolonged wounds are on the rise as a result of aging people, hence contributing to escalated expenditures within the healthcare system. The clinical evaluation and treatment of chronic wounds continue to pose challenges despite the advancement of different therapeutic approaches. This is mainly owing to the prolonged treatment duration and intricate processes involved in wound healing. Many conventional methods, such as the administration of growth factors, the use of wound dressings, and the application of skin grafts, are used to ease the process of wound healing across diverse wound types. Nevertheless, these therapeutic approaches may only be practical for some wounds, highlighting the need to advance alternative treatment modalities. Novel wound care technologies, such as nanotherapeutics, stem cell treatment, and 3D bioprinting, aim to improve therapeutic efficacy, prioritize skin regeneration, and minimize adverse effects. This review provides an updated overview of recent advancements in chronic wound healing and therapeutic management using innovative approaches.
Collapse
Affiliation(s)
- Abdullah Al Mamun
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, Zhejiang, China
| | - Chuxiao Shao
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, Zhejiang, China
| | - Peiwu Geng
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, Zhejiang, China
| | - Shuanghu Wang
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, Zhejiang, China
| | - Jian Xiao
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, Zhejiang, China
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- Department of Wound Healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
8
|
Shen X, Kong F, Wen J, Wang X, Huang C. The role of inflammation in central serous chorioretinopathy: From mechanisms to therapeutic prospects. Front Pharmacol 2024; 15:1200492. [PMID: 38835666 PMCID: PMC11148560 DOI: 10.3389/fphar.2024.1200492] [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: 04/05/2023] [Accepted: 05/06/2024] [Indexed: 06/06/2024] Open
Abstract
Central serous chorioretinopathy (CSC) is a leading cause of permanent vision loss, ranking fourth among macular diseases, trailing only age-related macular degeneration, diabetic retinopathy, and retinal vein obstruction. While mounting evidence implicates inflammation as a pivotal factor in the onset and advancement of CSC, the specific pathophysiological process and molecular mechanisms underlying inflammation remain incompletely understood. A complex network of cytokines, chemokines, and adhesion molecules interplay to trigger inflammatory and pathological cascades, highlighting the need for a comprehensive comprehension of the inflammation-related mechanisms behind CSC progression. In this piece, we examine the existing comprehension of CSC's pathology and pathogenesis. Additionally, we present an overview of the mechanisms underlying the onset and progression of CSC inflammation, followed by a thorough analysis and discussion of the potential of targeted inflammatory intervention for both preventing and treating CSC.
Collapse
Affiliation(s)
- Xiao Shen
- Department of Ophthalmology, Taizhou Central Hospital ( Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Fanhua Kong
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-Based Medical Materials, Wuhan, China
| | - Jing Wen
- Department of Ophthalmology, Taizhou Central Hospital ( Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Xiao Wang
- Department of Ophthalmology, Taizhou Central Hospital ( Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Chunlian Huang
- Department of Ophthalmology, Taizhou Central Hospital ( Taizhou University Hospital), Taizhou, Zhejiang, China
| |
Collapse
|
9
|
Liu J, He J, Liao Z, Chen X, Ye Y, Pang Q, Fan R. Environmental dose of 16 priority-controlled PAHs induce endothelial dysfunction: An in vivo and in vitro study. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 919:170711. [PMID: 38340817 DOI: 10.1016/j.scitotenv.2024.170711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 12/24/2023] [Accepted: 02/03/2024] [Indexed: 02/12/2024]
Abstract
Polycyclic aromatic hydrocarbons (PAHs) exposure is related to the occurrence of cardiovascular diseases (CVDs). Endothelial dysfunction is considered an initial event of CVDs. To confirm the relationship of PAHs exposure with endothelial dysfunction, 8-week-old male SD rats and primary human umbilical vein endothelial cells (HUVECs) were co-treated with environmental doses of 16 priority-controlled PAHs for 90 d and 48 h, respectively. Results showed that 10× PAHs exposure remarkably raised tumor necrosis factor-α and malonaldehyde levels in rat serum (p < 0.05), but had no effects on interleukin-8 levels and superoxide dismutase activity. The expressions of SIRT1 in HUVECs and rat aorta were attenuated after PAHs treatment. Interestingly, PAHs exposure did not activate the expression of total endothelial nitric oxide synthase (eNOS), but 10× PAHs exposure significantly elevated the expression of phosphorylated eNOS (Ser1177) in HUVECs and repressed it in aortas, accompanied with raised nitrite level both in serum and HUVECs by 48.50-253.70 %. PAHs exposure also led to the augment of endothelin-1 (ET-1) levels by 19.76-38.54 %, angiotensin (Ang II) levels by 20.09-39.69 % in HUVECs, but had no effects on ET-1 and Ang II levels in serum. Additionally, PAHs exposure improved endocan levels both in HUVECs and serum by 305.05-620.48 % and stimulated the THP-1 cells adhered to HUVECs (p < 0.05). After PAHs treatment, the smooth muscle alignment was disordered and the vascular smooth muscle locally proliferated in rat aorta. Notably, the systolic blood pressure of rats exposed to 10× PAHs increased significantly compared with the control ones (131.28 ± 5.20 vs 116.75 ± 5.33 mmHg). In summary, environmental chronic PAHs exposure may result in endothelial dysfunction in SD rats and primary HUVECs. Our research can confirm the cardiovascular damage caused by chronic exposure to PAHs and provide ideas for the prevention or intervention of CVDs affected by environmental factors.
Collapse
Affiliation(s)
- Jian Liu
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Jiaying He
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Zengquan Liao
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Xiaolin Chen
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Yufeng Ye
- Medical Imaging Institute of Panyu, Guangzhou 511486, China
| | - Qihua Pang
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Ruifang Fan
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Engineering Technology Research Center for Drug and Food Biological Resources Processing and Comprehensive Utilization, School of Life Sciences, South China Normal University, Guangzhou 510631, China.
| |
Collapse
|
10
|
Di Russo S, Liberati FR, Riva A, Di Fonzo F, Macone A, Giardina G, Arese M, Rinaldo S, Cutruzzolà F, Paone A. Beyond the barrier: the immune-inspired pathways of tumor extravasation. Cell Commun Signal 2024; 22:104. [PMID: 38331871 PMCID: PMC10851599 DOI: 10.1186/s12964-023-01429-1] [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: 11/06/2023] [Accepted: 12/08/2023] [Indexed: 02/10/2024] Open
Abstract
Extravasation is a fundamental step in the metastatic journey, where cancer cells exit the bloodstream and breach the endothelial cell barrier to infiltrate target tissues. The tactics cancer cells employ are sophisticated, closely reflecting those used by the immune system for tissue surveillance. Remarkably, tumor cells have been observed to form distinct associations or clusters with immune cells where neutrophils stand out as particularly crucial partners. These interactions are not accidental; they are critical for cancer cells to exploit the immune functions of neutrophils and successfully extravasate. In another strategy, tumor cells mimic the behavior and characteristics of immune cells. They release a suite of inflammatory mediators, which under normal circumstances, guide the processes of endothelium reshaping and facilitate the entry and movement of immune cells within tissues. In this review, we offer a new perspective on the tactics employed by cancer cells to extravasate and infiltrate target tissues. We delve into the myriad mechanisms that tumor cells borrow, adapt, and refine from the immune playbook. Video Abstract.
Collapse
Affiliation(s)
- Sara Di Russo
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti P.Le A. Moro 5, Rome, 00185, Italy
| | - Francesca Romana Liberati
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti P.Le A. Moro 5, Rome, 00185, Italy
| | - Agnese Riva
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti P.Le A. Moro 5, Rome, 00185, Italy
| | - Federica Di Fonzo
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti P.Le A. Moro 5, Rome, 00185, Italy
| | - Alberto Macone
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti P.Le A. Moro 5, Rome, 00185, Italy
| | - Giorgio Giardina
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti P.Le A. Moro 5, Rome, 00185, Italy
| | - Marzia Arese
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti P.Le A. Moro 5, Rome, 00185, Italy
| | - Serena Rinaldo
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti P.Le A. Moro 5, Rome, 00185, Italy
| | - Francesca Cutruzzolà
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti P.Le A. Moro 5, Rome, 00185, Italy
| | - Alessio Paone
- Department of Biochemical Sciences "Alessandro Rossi Fanelli", Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti P.Le A. Moro 5, Rome, 00185, Italy.
| |
Collapse
|
11
|
Vieceli Dalla Sega F, Fortini F, Licastro D, Monego SD, Degasperi M, Ascierto A, Marracino L, Severi P, D'Accolti M, Soffritti I, Brambilla M, Camera M, Tremoli E, Contoli M, Spadaro S, Campo G, Ferrari R, Caselli E, Rizzo P. Serum from COVID-19 patients promotes endothelial cell dysfunction through protease-activated receptor 2. Inflamm Res 2024; 73:117-130. [PMID: 38117300 DOI: 10.1007/s00011-023-01823-y] [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: 05/31/2023] [Revised: 10/06/2023] [Accepted: 11/13/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND Endothelial dysfunction plays a central role in the pathophysiology of COVID-19 and is closely linked to the severity and mortality of the disease. The inflammatory response to SARS-CoV-2 infection can alter the capacity of the endothelium to regulate vascular tone, immune responses, and the balance between anti-thrombotic and pro-thrombotic properties. However, the specific endothelial pathways altered during COVID-19 still need to be fully understood. OBJECTIVE In this study, we sought to identify molecular changes in endothelial cells induced by circulating factors characteristic of COVID-19. METHODS AND RESULTS To this aim, we cultured endothelial cells with sera from patients with COVID-19 or non-COVID-19 pneumonia. Through transcriptomic analysis, we were able to identify a distinctive endothelial phenotype that is induced by sera from COVID-19 patients. We confirmed and expanded this observation in vitro by showing that COVID-19 serum alters functional properties of endothelial cells leading to increased apoptosis, loss of barrier integrity, and hypercoagulability. Furthermore, we demonstrated that these endothelial dysfunctions are mediated by protease-activated receptor 2 (PAR-2), as predicted by transcriptome network analysis validated by in vitro functional assays. CONCLUSION Our findings provide the rationale for further studies to evaluate whether targeting PAR-2 may be a clinically effective strategy to counteract endothelial dysfunction in COVID-19.
Collapse
Affiliation(s)
| | | | | | | | | | - Alessia Ascierto
- Department of Translational Medicine and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Luisa Marracino
- Department of Translational Medicine and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Paolo Severi
- Department of Translational Medicine and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Maria D'Accolti
- Department of Chemical, Pharmaceutical, and Agricultural Sciences, and LTTA, Section of Microbiology, University of Ferrara, Ferrara, Italy
| | - Irene Soffritti
- Department of Chemical, Pharmaceutical, and Agricultural Sciences, and LTTA, Section of Microbiology, University of Ferrara, Ferrara, Italy
| | | | - Marina Camera
- Centro Cardiologico Monzino IRCCS, Milan, Italy
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan, Italy
| | - Elena Tremoli
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, Italy
| | - Marco Contoli
- Respiratory Section, Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Savino Spadaro
- Intensive Care Unit, Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Gianluca Campo
- Cardiology Unit, Azienda Ospedaliero-Universitaria di Ferrara, University of Ferrara, Ferrara, Italy
| | - Roberto Ferrari
- Department of Translational Medicine and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Elisabetta Caselli
- Department of Chemical, Pharmaceutical, and Agricultural Sciences, and LTTA, Section of Microbiology, University of Ferrara, Ferrara, Italy
| | - Paola Rizzo
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, Italy
- Department of Translational Medicine and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| |
Collapse
|
12
|
Ferreira LB, Williams KA, Best G, Haydinger CD, Smith JR. Inflammatory cytokines as mediators of retinal endothelial barrier dysfunction in non-infectious uveitis. Clin Transl Immunology 2023; 12:e1479. [PMID: 38090668 PMCID: PMC10714664 DOI: 10.1002/cti2.1479] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 09/22/2023] [Accepted: 11/21/2023] [Indexed: 06/30/2024] Open
Abstract
Characterised by intraocular inflammation, non-infectious uveitis includes a large group of autoimmune and autoinflammatory diseases that either involve the eye alone or have both ocular and systemic manifestations. When non-infectious uveitis involves the posterior segment of the eye, specifically the retina, there is substantial risk of vision loss, often linked to breakdown of the inner blood-retinal barrier. This barrier is formed by non-fenestrated retinal vascular endothelial cells, reinforced by supporting cells that include pericytes, Müller cells and astrocytes. Across the published literature, a group of inflammatory cytokines stand out as prominent mediators of intraocular inflammation, with effects on the retinal endothelium that may contribute to breakdown of the inner blood-retinal barrier, namely tumour necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, IL-8, IL-17 and chemokine C-C motif ligand (CCL)2. This article reviews the function of each cytokine and discusses the evidence for their involvement in retinal endothelial barrier dysfunction in non-infectious uveitis, including basic laboratory investigations, studies of ocular fluids collected from patients with non-infectious uveitis, and results of clinical treatment trials. The review also outlines gaps in knowledge in this area. Understanding the disease processes at a molecular level can suggest treatment alternatives that are directed against appropriate biological targets to protect the posterior segment of eye and preserve vision in non-infectious uveitis.
Collapse
Affiliation(s)
| | - Keryn A Williams
- Flinders University College of Medicine and Public HealthAdelaideSAAustralia
| | - Giles Best
- Flinders University College of Medicine and Public HealthAdelaideSAAustralia
| | - Cameron D Haydinger
- Flinders University College of Medicine and Public HealthAdelaideSAAustralia
| | - Justine R Smith
- Flinders University College of Medicine and Public HealthAdelaideSAAustralia
| |
Collapse
|
13
|
Colás-Algora N, Muñoz-Pinillos P, Cacho-Navas C, Avendaño-Ortiz J, de Rivas G, Barroso S, López-Collazo E, Millán J. Simultaneous Targeting of IL-1-Signaling and IL-6-Trans-Signaling Preserves Human Pulmonary Endothelial Barrier Function During a Cytokine Storm-Brief Report. Arterioscler Thromb Vasc Biol 2023; 43:2213-2222. [PMID: 37732482 DOI: 10.1161/atvbaha.123.319695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 09/06/2023] [Indexed: 09/22/2023]
Abstract
BACKGROUND Systemic inflammatory diseases, such as sepsis and severe COVID-19, provoke acute respiratory distress syndrome in which the pathological hyperpermeability of the microvasculature, induced by uncontrolled inflammatory stimulation, causes pulmonary edema. Identifying the inflammatory mediators that induce human lung microvascular endothelial cell barrier dysfunction is essential to find the best anti-inflammatory treatments for critically ill acute respiratory distress syndrome patients. METHODS We have compared the responses of primary human lung microvascular endothelial cells to the main inflammatory mediators involved in cytokine storms induced by sepsis and SARS-CoV2 pulmonary infection and to sera from healthy donors and severely ill patients with sepsis. Endothelial barrier function was measured by electric cell-substrate impedance sensing, quantitative confocal microscopy, and Western blot. RESULTS The human lung microvascular endothelial cell barrier was completely disrupted by IL (interleukin)-6 conjugated with soluble IL-6R (IL-6 receptor) and by IL-1β (interleukin-1beta), moderately affected by TNF (tumor necrosis factor)-α and IFN (interferon)-γ and unaffected by other cytokines and chemokines, such as IL-6, IL-8, MCP (monocyte chemoattractant protein)-1 and MCP-3. The inhibition of IL-1 and IL-6R simultaneously, but not separately, significantly reduced endothelial hyperpermeability on exposing human lung microvascular endothelial cells to a cytokine storm consisting of 8 inflammatory mediators or to sera from patients with sepsis. Simultaneous inhibition of IL-1 and JAK (Janus kinase)-STAT (signal transducer and activator of transcription protein), a signaling node downstream IL-6 and IFN-γ, also prevented septic serum-induced endothelial barrier disruption. CONCLUSIONS These findings strongly suggest a major role for both IL-6 trans-signaling and IL-1β signaling in the pathological increase in permeability of the human lung microvasculature and reveal combinatorial strategies that enable the gradual control of pulmonary endothelial barrier function in response to a cytokine storm.
Collapse
Affiliation(s)
- Natalia Colás-Algora
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Cantoblanco, Madrid, Spain (N.C.-A., P.M.-P., C.C.-N., G.d.R., S.B., J.M.)
| | - Pablo Muñoz-Pinillos
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Cantoblanco, Madrid, Spain (N.C.-A., P.M.-P., C.C.-N., G.d.R., S.B., J.M.)
| | - Cristina Cacho-Navas
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Cantoblanco, Madrid, Spain (N.C.-A., P.M.-P., C.C.-N., G.d.R., S.B., J.M.)
| | - José Avendaño-Ortiz
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain (J.A.O., E.L.-C.)
- CIBER of Respiratory Diseases (CIBERES), Madrid, Spain (J.A.O., E.L.-C.)
| | - Gema de Rivas
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Cantoblanco, Madrid, Spain (N.C.-A., P.M.-P., C.C.-N., G.d.R., S.B., J.M.)
| | - Susana Barroso
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Cantoblanco, Madrid, Spain (N.C.-A., P.M.-P., C.C.-N., G.d.R., S.B., J.M.)
| | - Eduardo López-Collazo
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain (J.A.O., E.L.-C.)
- CIBER of Respiratory Diseases (CIBERES), Madrid, Spain (J.A.O., E.L.-C.)
| | - Jaime Millán
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Cantoblanco, Madrid, Spain (N.C.-A., P.M.-P., C.C.-N., G.d.R., S.B., J.M.)
| |
Collapse
|
14
|
Wittig F, Pannenberg L, Schwarz R, Bekeschus S, Ramer R, Hinz B. Antiangiogenic Action of JZL184 on Endothelial Cells via Inhibition of VEGF Expression in Hypoxic Lung Cancer Cells. Cells 2023; 12:2332. [PMID: 37830546 PMCID: PMC10572003 DOI: 10.3390/cells12192332] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/16/2023] [Accepted: 08/25/2023] [Indexed: 10/14/2023] Open
Abstract
JZL184, an inhibitor of monoacylglycerol lipase (MAGL) and thus of the degradation of the endocannabinoid 2-arachidonoylglycerol (2-AG), mediates various anticancer effects in preclinical studies. However, studies on the effect of this or other MAGL inhibitors under hypoxia, an important factor in tumor biology and response to cancer therapy, have not yet been performed in cancer cells. In the present study, the impact of the conditioned media (CM) of A549 and H358 lung cancer cells incubated with JZL184 under hypoxic conditions on the angiogenic properties of human umbilical vein endothelial cells (HUVECs) was investigated. Treatment of HUVECs with CM derived from cancer cells cultured for 48 h under hypoxic conditions was associated with a substantial increase in migration and tube formation compared with unconditioned medium, which was inhibited when cancer cells were incubated with JZL184. In this process, JZL184 led to a significant increase in 2-AG levels in both cell lines. Analysis of a panel of proangiogenic factors revealed inhibition of hypoxia-induced vascular endothelial growth factor (VEGF) expression by JZL184. Antiangiogenic and VEGF-lowering effects were also demonstrated for the MAGL inhibitor MJN110. Receptor antagonist experiments suggest partial involvement of the cannabinoid receptors CB1 and CB2 in the antiangiogenic and VEGF-lowering effects induced by JZL184. The functional importance of VEGF for angiogenesis in the selected system is supported by observations showing inhibition of VEGF receptor 2 (VEGFR2) phosphorylation in HUVECs by CM from hypoxic cancer cells treated with JZL184 or when hypoxic cancer cell-derived CM was spiked with a neutralizing VEGF antibody. On the other hand, JZL184 did not exert a direct effect on VEGFR2 activation induced by recombinant VEGF, so there seems to be no downstream effect on already released VEGF. In conclusion, these results reveal a novel mechanism of antiangiogenic action of JZL184 under conditions of hypoxic tumor-endothelial communication.
Collapse
Affiliation(s)
- Felix Wittig
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, Schillingallee 70, 18057 Rostock, Germany; (F.W.); (L.P.); (R.S.); (R.R.)
| | - Liza Pannenberg
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, Schillingallee 70, 18057 Rostock, Germany; (F.W.); (L.P.); (R.S.); (R.R.)
| | - Rico Schwarz
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, Schillingallee 70, 18057 Rostock, Germany; (F.W.); (L.P.); (R.S.); (R.R.)
| | - Sander Bekeschus
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany;
| | - Robert Ramer
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, Schillingallee 70, 18057 Rostock, Germany; (F.W.); (L.P.); (R.S.); (R.R.)
| | - Burkhard Hinz
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, Schillingallee 70, 18057 Rostock, Germany; (F.W.); (L.P.); (R.S.); (R.R.)
| |
Collapse
|
15
|
Goh I, Botting RA, Rose A, Webb S, Engelbert J, Gitton Y, Stephenson E, Londoño MQ, Mather M, Mende N, Imaz-Rosshandler I, Yang L, Horsfall D, Basurto-Lozada D, Chipampe NJ, Rook V, Lee JTH, Ton ML, Keitley D, Mazin P, Vijayabaskar M, Hannah R, Gambardella L, Green K, Ballereau S, Inoue M, Tuck E, Lorenzi V, Kwakwa K, Alsinet C, Olabi B, Miah M, Admane C, Popescu DM, Acres M, Dixon D, Ness T, Coulthard R, Lisgo S, Henderson DJ, Dann E, Suo C, Kinston SJ, Park JE, Polanski K, Marioni J, van Dongen S, Meyer KB, de Bruijn M, Palis J, Behjati S, Laurenti E, Wilson NK, Vento-Tormo R, Chédotal A, Bayraktar O, Roberts I, Jardine L, Göttgens B, Teichmann SA, Haniffa M. Yolk sac cell atlas reveals multiorgan functions during human early development. Science 2023; 381:eadd7564. [PMID: 37590359 PMCID: PMC7614978 DOI: 10.1126/science.add7564] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 07/03/2023] [Indexed: 08/19/2023]
Abstract
The extraembryonic yolk sac (YS) ensures delivery of nutritional support and oxygen to the developing embryo but remains ill-defined in humans. We therefore assembled a comprehensive multiomic reference of the human YS from 3 to 8 postconception weeks by integrating single-cell protein and gene expression data. Beyond its recognized role as a site of hematopoiesis, we highlight roles in metabolism, coagulation, vascular development, and hematopoietic regulation. We reconstructed the emergence and decline of YS hematopoietic stem and progenitor cells from hemogenic endothelium and revealed a YS-specific accelerated route to macrophage production that seeds developing organs. The multiorgan functions of the YS are superseded as intraembryonic organs develop, effecting a multifaceted relay of vital functions as pregnancy proceeds.
Collapse
Affiliation(s)
- Issac Goh
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton,
Cambridge CB10 1SA, UK
- Biosciences Institute, Newcastle University, NE2 4HH, UK
| | - Rachel A. Botting
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton,
Cambridge CB10 1SA, UK
- Biosciences Institute, Newcastle University, NE2 4HH, UK
| | - Antony Rose
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton,
Cambridge CB10 1SA, UK
- Biosciences Institute, Newcastle University, NE2 4HH, UK
| | - Simone Webb
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton,
Cambridge CB10 1SA, UK
- Biosciences Institute, Newcastle University, NE2 4HH, UK
| | | | - Yorick Gitton
- Sorbonne Université, INSERM, CNRS, Institut de la Vision,
Paris, France
| | - Emily Stephenson
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton,
Cambridge CB10 1SA, UK
- Biosciences Institute, Newcastle University, NE2 4HH, UK
| | | | - Michael Mather
- Biosciences Institute, Newcastle University, NE2 4HH, UK
| | - Nicole Mende
- Department of Haematology, Wellcome-MRC Cambridge Stem Cell
Institute, CB2 0AW, UK
| | - Ivan Imaz-Rosshandler
- Department of Haematology, Wellcome-MRC Cambridge Stem Cell
Institute, CB2 0AW, UK
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus,
CD2 0QH, UK
| | - Lu Yang
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton,
Cambridge CB10 1SA, UK
| | - Dave Horsfall
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton,
Cambridge CB10 1SA, UK
- Biosciences Institute, Newcastle University, NE2 4HH, UK
| | - Daniela Basurto-Lozada
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton,
Cambridge CB10 1SA, UK
- Biosciences Institute, Newcastle University, NE2 4HH, UK
| | - Nana-Jane Chipampe
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton,
Cambridge CB10 1SA, UK
| | - Victoria Rook
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton,
Cambridge CB10 1SA, UK
| | - Jimmy Tsz Hang Lee
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton,
Cambridge CB10 1SA, UK
| | - Mai-Linh Ton
- Department of Haematology, Wellcome-MRC Cambridge Stem Cell
Institute, CB2 0AW, UK
| | - Daniel Keitley
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton,
Cambridge CB10 1SA, UK
- Department of Zoology, University of Cambridge, Cambridge UK
| | - Pavel Mazin
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton,
Cambridge CB10 1SA, UK
| | - M.S. Vijayabaskar
- Department of Haematology, Wellcome-MRC Cambridge Stem Cell
Institute, CB2 0AW, UK
| | - Rebecca Hannah
- Department of Haematology, Wellcome-MRC Cambridge Stem Cell
Institute, CB2 0AW, UK
| | - Laure Gambardella
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton,
Cambridge CB10 1SA, UK
| | - Kile Green
- Translational and Clinical Research Institute, Newcastle University,
NE2 4HH, UK
| | - Stephane Ballereau
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton,
Cambridge CB10 1SA, UK
| | - Megumi Inoue
- Sorbonne Université, INSERM, CNRS, Institut de la Vision,
Paris, France
| | - Elizabeth Tuck
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton,
Cambridge CB10 1SA, UK
| | - Valentina Lorenzi
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton,
Cambridge CB10 1SA, UK
| | - Kwasi Kwakwa
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton,
Cambridge CB10 1SA, UK
| | - Clara Alsinet
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton,
Cambridge CB10 1SA, UK
- Centre Nacional d’Analisi Genomica-Centre de Regulacio
Genomica (CNAG-CRG), Barcelona Institute of Science and Technology (BIST),
Barcelona, Spain
| | - Bayanne Olabi
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton,
Cambridge CB10 1SA, UK
- Biosciences Institute, Newcastle University, NE2 4HH, UK
| | - Mohi Miah
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton,
Cambridge CB10 1SA, UK
- Biosciences Institute, Newcastle University, NE2 4HH, UK
| | - Chloe Admane
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton,
Cambridge CB10 1SA, UK
- Biosciences Institute, Newcastle University, NE2 4HH, UK
| | | | - Meghan Acres
- Biosciences Institute, Newcastle University, NE2 4HH, UK
| | - David Dixon
- Biosciences Institute, Newcastle University, NE2 4HH, UK
| | - Thomas Ness
- NovoPath, Department of Pathology, Newcastle Hospitals NHS
Foundation Trust, Newcastle upon Tyne, UK
| | - Rowen Coulthard
- NovoPath, Department of Pathology, Newcastle Hospitals NHS
Foundation Trust, Newcastle upon Tyne, UK
| | - Steven Lisgo
- Biosciences Institute, Newcastle University, NE2 4HH, UK
| | | | - Emma Dann
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton,
Cambridge CB10 1SA, UK
| | - Chenqu Suo
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton,
Cambridge CB10 1SA, UK
| | - Sarah J. Kinston
- Department of Haematology, Wellcome-MRC Cambridge Stem Cell
Institute, CB2 0AW, UK
| | - Jong-eun Park
- Korea Advanced Institute of Science and Technology, Daejeon, South
Korea
| | - Krzysztof Polanski
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton,
Cambridge CB10 1SA, UK
| | - John Marioni
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton,
Cambridge CB10 1SA, UK
- EMBL-EBI, Wellcome Genome Campus, Cambridge, UK
- CRUK Cambridge Institute, University of Cambridge, Cambridge,
UK
| | - Stijn van Dongen
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton,
Cambridge CB10 1SA, UK
| | - Kerstin B. Meyer
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton,
Cambridge CB10 1SA, UK
| | - Marella de Bruijn
- MRC Molecular Haematology Unit, MRC Weatherall Institute of
Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, OX3 9DS,
UK
| | - James Palis
- Department of Pediatrics, University of Rochester Medical Center,
Rochester, 14642, NY, USA
| | - Sam Behjati
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton,
Cambridge CB10 1SA, UK
- Department of Paediatrics, University of Cambridge, Cambridge,
UK
| | - Elisa Laurenti
- Department of Haematology, Wellcome-MRC Cambridge Stem Cell
Institute, CB2 0AW, UK
| | - Nicola K. Wilson
- Department of Haematology, Wellcome-MRC Cambridge Stem Cell
Institute, CB2 0AW, UK
| | - Roser Vento-Tormo
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton,
Cambridge CB10 1SA, UK
| | - Alain Chédotal
- Sorbonne Université, INSERM, CNRS, Institut de la Vision,
Paris, France
| | - Omer Bayraktar
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton,
Cambridge CB10 1SA, UK
| | - Irene Roberts
- Department of Paediatrics, University of Oxford, OX3 9DS, UK
| | - Laura Jardine
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton,
Cambridge CB10 1SA, UK
- Biosciences Institute, Newcastle University, NE2 4HH, UK
| | - Berthold Göttgens
- Department of Haematology, Wellcome-MRC Cambridge Stem Cell
Institute, CB2 0AW, UK
| | - Sarah A. Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton,
Cambridge CB10 1SA, UK
- Theory of Condensed Matter Group, Cavendish Laboratory/Department
of Physics, University of Cambridge, Cambridge, UK
| | - Muzlifah Haniffa
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton,
Cambridge CB10 1SA, UK
- Biosciences Institute, Newcastle University, NE2 4HH, UK
- Department of Dermatology and NIHR Newcastle Biomedical Research
Centre, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, NE1 4LP,
UK
| |
Collapse
|
16
|
Eom Y, Eom SY, Lee J, Hwang S, Won J, Kim H, Chung S, Kim HJ, Lee MY. Therapeutic Effects and Underlying Mechanism of SOCS-com Gene-Transfected ADMSCs in Pressure Ulcer Mouse Models. Cells 2023; 12:1840. [PMID: 37508509 PMCID: PMC10378383 DOI: 10.3390/cells12141840] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Although the proportion of ulcer patients with medical problems among the elderly has increased with the extension of human life expectancy, treatment efficiency is drastically low, incurring substantial social costs. MSCs have independent regeneration potential, making them useful in clinical trials of difficult-to-treat diseases. In particular, ADMSCs are promising in the stem cell therapy industry as they can be obtained in vast amounts using non-invasive methods. Furthermore, studies are underway to enhance the regeneration potential of ADMSCs using cytokines, growth factors, and gene delivery to generate highly functional ADMSCs. In this study, key regulators of wound healing, SOCS-1, -3, and -5, were combined to maximize the regenerative potential of ADMSCs in pressure ulcer treatments. After transfecting SOCS-1, -3, -5, and SOCS-com into ADMSCs using a non-viral method, the expression of the inflammatory factors TNF-alpha, INF-gamma, and IL-10 was confirmed. ADMSCs transfected with SOCS-com showed decreased overall expression of inflammatory factors and increased expression of anti-inflammatory factors. Based on these results, we implanted ADMSCs transfected with SOCS-com into a pressure ulcer mouse model to observe their subsequent wound-healing effects. Notably, SOCS-com improved wound closure in ulcers, and reconstruction of the epidermis and dermis was observed. The healing mechanism of ADMSCs transfected with SOCS-com was examined by RNA sequencing. Gene analysis results confirmed that expression changes occurred in genes of key regulators of wound healing, such as chemokines, MMP-1, 9, CSF-2, and IL-33, and that such genetic changes enhanced wound healing in ulcers. Based on these results, we demonstrate the potential of ADMSCs transfected with SOCS-com as an ulcer treatment tool.
Collapse
Affiliation(s)
- Youngsic Eom
- Department of Medical Science, College of Medical Sciences, Soonchunhyang University, Asan 31538, Republic of Korea
| | - So Young Eom
- School of Mechanical Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Jeonghwa Lee
- Department of Medical Science, College of Medical Sciences, Soonchunhyang University, Asan 31538, Republic of Korea
| | - Saeyeon Hwang
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 34943, Republic of Korea
| | - Jihee Won
- School of Mechanical Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Hyunsoo Kim
- School of Mechanical Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Seok Chung
- School of Mechanical Engineering, Korea University, Seoul 02841, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
- Center for Brain Technology, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Hye Joung Kim
- Institute of Chemical Engineering Convergence System, Korea University, Seoul 02841, Republic of Korea
| | - Mi-Young Lee
- Department of Medical Science, College of Medical Sciences, Soonchunhyang University, Asan 31538, Republic of Korea
| |
Collapse
|
17
|
de Rus Jacquet A, Alpaugh M, Denis HL, Tancredi JL, Boutin M, Decaestecker J, Beauparlant C, Herrmann L, Saint-Pierre M, Parent M, Droit A, Breton S, Cicchetti F. The contribution of inflammatory astrocytes to BBB impairments in a brain-chip model of Parkinson's disease. Nat Commun 2023; 14:3651. [PMID: 37339976 DOI: 10.1038/s41467-023-39038-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 05/26/2023] [Indexed: 06/22/2023] Open
Abstract
Astrocyte dysfunction has previously been linked to multiple neurodegenerative disorders including Parkinson's disease (PD). Among their many roles, astrocytes are mediators of the brain immune response, and astrocyte reactivity is a pathological feature of PD. They are also involved in the formation and maintenance of the blood-brain barrier (BBB), but barrier integrity is compromised in people with PD. This study focuses on an unexplored area of PD pathogenesis by characterizing the interplay between astrocytes, inflammation and BBB integrity, and by combining patient-derived induced pluripotent stem cells with microfluidic technologies to generate a 3D human BBB chip. Here we report that astrocytes derived from female donors harboring the PD-related LRRK2 G2019S mutation are pro-inflammatory and fail to support the formation of a functional capillary in vitro. We show that inhibition of MEK1/2 signaling attenuates the inflammatory profile of mutant astrocytes and rescues BBB formation, providing insights into mechanisms regulating barrier integrity in PD. Lastly, we confirm that vascular changes are also observed in the human postmortem substantia nigra of both males and females with PD.
Collapse
Affiliation(s)
- A de Rus Jacquet
- Centre de Recherche du CHU de Québec - Université Laval, Axe Neurosciences, Québec, QC, G1V 4G2, Canada.
- Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC, G1V 0A6, Canada.
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, 20147, USA.
| | - M Alpaugh
- Centre de Recherche du CHU de Québec - Université Laval, Axe Neurosciences, Québec, QC, G1V 4G2, Canada
- Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC, G1V 0A6, Canada
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - H L Denis
- Centre de Recherche du CHU de Québec - Université Laval, Axe Neurosciences, Québec, QC, G1V 4G2, Canada
- Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC, G1V 0A6, Canada
| | - J L Tancredi
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, 20147, USA
- Cell Biology R&D, Thermo Fisher Scientific, Frederick, MD, 21704, USA
| | - M Boutin
- Centre de Recherche du CHU de Québec - Université Laval, Axe Neurosciences, Québec, QC, G1V 4G2, Canada
| | - J Decaestecker
- Centre de Recherche du CHU de Québec - Université Laval, Axe Endocrinologie et Néphrologie, Québec, QC, G1V 4G2, Canada
| | - C Beauparlant
- Centre de Recherche du CHU de Québec - Université Laval, Axe Endocrinologie et Néphrologie, Québec, QC, G1V 4G2, Canada
| | - L Herrmann
- Centre de Recherche du CHU de Québec - Université Laval, Axe Endocrinologie et Néphrologie, Québec, QC, G1V 4G2, Canada
| | - M Saint-Pierre
- Centre de Recherche du CHU de Québec - Université Laval, Axe Neurosciences, Québec, QC, G1V 4G2, Canada
| | - M Parent
- Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC, G1V 0A6, Canada
- CERVO Brain Research Center, Québec, QC, G1E 1T2, Canada
| | - A Droit
- Centre de Recherche du CHU de Québec - Université Laval, Axe Endocrinologie et Néphrologie, Québec, QC, G1V 4G2, Canada
| | - S Breton
- Centre de Recherche du CHU de Québec - Université Laval, Axe Reproduction, santé de la mère et de l'enfant, Québec, QC, G1V 4G2, Canada
- Centre de recherche en reproduction, développement et santé intergénérationnelle, Université Laval, Québec, QC, G1V 4G2, Canada
| | - F Cicchetti
- Centre de Recherche du CHU de Québec - Université Laval, Axe Neurosciences, Québec, QC, G1V 4G2, Canada.
- Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC, G1V 0A6, Canada.
| |
Collapse
|
18
|
Zhou H, Tu LN, Giachelli C, Nigam V, Scatena M. Monocyte Adhesion and Transmigration Through Endothelium Following Cardiopulmonary Bypass Shearing is Mediated by IL-8 Signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.05.543811. [PMID: 37333089 PMCID: PMC10274614 DOI: 10.1101/2023.06.05.543811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
BackgroundThe use of cardiopulmonary bypass (CPB) can induce sterile systemic inflammation that contributes to morbidity and mortality, especially in children. Patients have been found to have increased expression of cytokines and transmigration of leukocytes during and after CPB. Previous work has demonstrated that the supraphysiologic shear stresses present during CPB are sufficient to induce proinflammatory behavior in non-adherent monocytes. The interactions between shear stimulated monocytes and vascular endothelial cells have not been well studied and have important translational implications.MethodsTo test the hypothesis that non-physiological shear stress experienced by monocytes during CPB affects the integrity and function of the endothelial monolayer via IL-8 signaling pathway, we have used an in vitro CPB model to study the interaction between THP-1 monocyte-like cells and human neonatal dermal microvascular endothelial cells (HNDMVECs). THP-1 cells were sheared in polyvinyl chloride (PVC) tubing at 2.1 Pa, twice of physiological shear stress, for 2 hours. Interactions between THP-1 cells and HNDMVECs were characterized after coculture.ResultsWe found that sheared THP-1 cells adhered to and transmigrated through the HNDMVEC monolayer more readily than static controls. When co-culturing, sheared THP-1 cells also disrupted in the VE-cadherin and led to reorganization of cytoskeletal F-actin of HNDMVECs. Treating HNDMVECs with IL-8 resulted in upregulation of vascular cell adhesion molecule 1 (VCAM-1) and intercellular adhesion molecule 1 (ICAM-1) while also increasing the adherence of non-sheared THP-1 cells. Preincubating HNDMVECs with Reparixin, an inhibitor of CXCR2/IL-8 receptor inhibited sheared THP-1 cell adhesion to the HNDMVECs.ConclusionsThese results suggested that IL-8 not only increases the endothelium permeability during monocyte migration, but also affects the initial adhesion of monocytes in a CPB setup. This study revealed a novel mechanism of post-CPB inflammation and will contribute to the development of targeted therapeutics to prevent and repair the damage to neonatal patients.HighlightsShear stress in a CPB-like environment promoted the adhesion and transmigration of monocytes to and through endothelial monolayer.Treating endothelial monolayer with sheared monocytes led to disruption of VE-cadherin and reorganization of F-actin.Interaction between sheared monocytes resulted in a significant increase of IL-8 release.Inhibiting IL-8 receptor prevented sheared monocyte adhesion, while IL-8 promoted naive monocyte adhesion.
Collapse
Affiliation(s)
- Hao Zhou
- University of Washington, Seattle, WA
| | - Lan N Tu
- Seattle Children's Hospital, Seattle, WA
| | | | - Vishal Nigam
- University of Washington, Seattle, WA
- Seattle Children's Hospital, Seattle, WA
| | | |
Collapse
|
19
|
Ruze R, Song J, Yin X, Chen Y, Xu R, Wang C, Zhao Y. Mechanisms of obesity- and diabetes mellitus-related pancreatic carcinogenesis: a comprehensive and systematic review. Signal Transduct Target Ther 2023; 8:139. [PMID: 36964133 PMCID: PMC10039087 DOI: 10.1038/s41392-023-01376-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 01/31/2023] [Accepted: 02/15/2023] [Indexed: 03/26/2023] Open
Abstract
Research on obesity- and diabetes mellitus (DM)-related carcinogenesis has expanded exponentially since these two diseases were recognized as important risk factors for cancers. The growing interest in this area is prominently actuated by the increasing obesity and DM prevalence, which is partially responsible for the slight but constant increase in pancreatic cancer (PC) occurrence. PC is a highly lethal malignancy characterized by its insidious symptoms, delayed diagnosis, and devastating prognosis. The intricate process of obesity and DM promoting pancreatic carcinogenesis involves their local impact on the pancreas and concurrent whole-body systemic changes that are suitable for cancer initiation. The main mechanisms involved in this process include the excessive accumulation of various nutrients and metabolites promoting carcinogenesis directly while also aggravating mutagenic and carcinogenic metabolic disorders by affecting multiple pathways. Detrimental alterations in gastrointestinal and sex hormone levels and microbiome dysfunction further compromise immunometabolic regulation and contribute to the establishment of an immunosuppressive tumor microenvironment (TME) for carcinogenesis, which can be exacerbated by several crucial pathophysiological processes and TME components, such as autophagy, endoplasmic reticulum stress, oxidative stress, epithelial-mesenchymal transition, and exosome secretion. This review provides a comprehensive and critical analysis of the immunometabolic mechanisms of obesity- and DM-related pancreatic carcinogenesis and dissects how metabolic disorders impair anticancer immunity and influence pathophysiological processes to favor cancer initiation.
Collapse
Affiliation(s)
- Rexiati Ruze
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- Key Laboratory of Research in Pancreatic Tumors, Chinese Academy of Medical Sciences, 100023, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, No. 9 Dongdan Santiao, Beijing, China
| | - Jianlu Song
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- Key Laboratory of Research in Pancreatic Tumors, Chinese Academy of Medical Sciences, 100023, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, No. 9 Dongdan Santiao, Beijing, China
| | - Xinpeng Yin
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- Key Laboratory of Research in Pancreatic Tumors, Chinese Academy of Medical Sciences, 100023, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, No. 9 Dongdan Santiao, Beijing, China
| | - Yuan Chen
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- Key Laboratory of Research in Pancreatic Tumors, Chinese Academy of Medical Sciences, 100023, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, No. 9 Dongdan Santiao, Beijing, China
| | - Ruiyuan Xu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- Key Laboratory of Research in Pancreatic Tumors, Chinese Academy of Medical Sciences, 100023, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, No. 9 Dongdan Santiao, Beijing, China
| | - Chengcheng Wang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China.
- Key Laboratory of Research in Pancreatic Tumors, Chinese Academy of Medical Sciences, 100023, Beijing, China.
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China.
- Key Laboratory of Research in Pancreatic Tumors, Chinese Academy of Medical Sciences, 100023, Beijing, China.
| |
Collapse
|
20
|
Habanjar O, Bingula R, Decombat C, Diab-Assaf M, Caldefie-Chezet F, Delort L. Crosstalk of Inflammatory Cytokines within the Breast Tumor Microenvironment. Int J Mol Sci 2023; 24:4002. [PMID: 36835413 PMCID: PMC9964711 DOI: 10.3390/ijms24044002] [Citation(s) in RCA: 71] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/10/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
Several immune and immunocompetent cells, including dendritic cells, macrophages, adipocytes, natural killer cells, T cells, and B cells, are significantly correlated with the complex discipline of oncology. Cytotoxic innate and adaptive immune cells can block tumor proliferation, and others can prevent the immune system from rejecting malignant cells and provide a favorable environment for tumor progression. These cells communicate with the microenvironment through cytokines, a chemical messenger, in an endocrine, paracrine, or autocrine manner. These cytokines play an important role in health and disease, particularly in host immune responses to infection and inflammation. They include chemokines, interleukins (ILs), adipokines, interferons, colony-stimulating factors (CSFs), and tumor necrosis factor (TNF), which are produced by a wide range of cells, including immune cells, such as macrophages, B-cells, T-cells, and mast cells, as well as endothelial cells, fibroblasts, a variety of stromal cells, and some cancer cells. Cytokines play a crucial role in cancer and cancer-related inflammation, with direct and indirect effects on tumor antagonistic or tumor promoting functions. They have been extensively researched as immunostimulatory mediators to promote the generation, migration and recruitment of immune cells that contribute to an effective antitumor immune response or pro-tumor microenvironment. Thus, in many cancers such as breast cancer, cytokines including leptin, IL-1B, IL-6, IL-8, IL-23, IL-17, and IL-10 stimulate while others including IL-2, IL-12, and IFN-γ, inhibit cancer proliferation and/or invasion and enhance the body's anti-tumor defense. Indeed, the multifactorial functions of cytokines in tumorigenesis will advance our understanding of cytokine crosstalk pathways in the tumor microenvironment, such as JAK/STAT, PI3K, AKT, Rac, MAPK, NF-κB, JunB, cFos, and mTOR, which are involved in angiogenesis, cancer proliferation and metastasis. Accordingly, targeting and blocking tumor-promoting cytokines or activating and amplifying tumor-inhibiting cytokines are considered cancer-directed therapies. Here, we focus on the role of the inflammatory cytokine system in pro- and anti-tumor immune responses, discuss cytokine pathways involved in immune responses to cancer and some anti-cancer therapeutic applications.
Collapse
Affiliation(s)
- Ola Habanjar
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France
| | - Rea Bingula
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France
| | - Caroline Decombat
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France
| | - Mona Diab-Assaf
- Equipe Tumorigénèse Pharmacologie Moléculaire et Anticancéreuse, Faculté des Sciences II, Université Libanaise Fanar, Beyrouth 1500, Lebanon
| | - Florence Caldefie-Chezet
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France
| | - Laetitia Delort
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France
| |
Collapse
|
21
|
Zhang Z, Tan J, Jin W, Qian H, Wang L, Zhou H, Yuan Y, Wu X. Severe fever with thrombocytopenia syndrome virus trends and hotspots in clinical research: A bibliometric analysis of global research. Front Public Health 2023; 11:1120462. [PMID: 36817929 PMCID: PMC9933999 DOI: 10.3389/fpubh.2023.1120462] [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: 12/10/2022] [Accepted: 01/18/2023] [Indexed: 02/05/2023] Open
Abstract
Background Since severe fever with thrombocytopenia syndrome virus (SFTSV) was first reported in 2009, a large number of relevant studies have been published. However, no bibliometrics analysis has been conducted on the literature focusing on SFTSV. This study aims to evaluate the research hotspots and future development trends of SFTSV research through bibliometric analysis, and to provide a new perspective and reference for future SFTSV research and the prevention of SFTSV. Methods We retrieved global publications on SFTSV from the Web of Science Core Collection (WoSCC) and Scopus databases from inception of the database until 2022 using VOSviewer software and CiteSpace was used for bibliometric analysis. Results The number of SFTSV-related publications has increased rapidly since 2011, peaking in 2021. A total of 45 countries/regions have published relevant publications, with China topping the list with 359. The Viruses-Basel has published the most papers on SFTSV. In addition, Yu et al. have made the greatest contribution to SFTSV research, with their published paper being the most frequently cited. The most popular SFTSV study topics included: (1) pathogenesis and symptoms, (2) characteristics of the virus and infected patients, and (3) transmission mechanism and risk factors for SFTSV. Conclusions In this study, we provide a detailed description of the research developments in SFTSV since its discovery and summarize the SFTSV research trends. SFTSV research is in a phase of explosive development, and a large number of publications have been published in the past decade. There is a lack of collaboration between countries and institutions, and international collaboration and exchanges should be strengthened in the future. The current research hotpots of SFTSV is antiviral therapy, immunotherapy, virus transmission mechanism and immune response.
Collapse
Affiliation(s)
- Zhengyu Zhang
- Medical Records Department, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Juntao Tan
- Operation Management Office, Affiliated Banan Hospital of Chongqing Medical University, Chongqing, China
| | - Wen Jin
- Medical Records Department, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hong Qian
- Medical Records Department, The First Hospital of Lanzhou University, Lanzhou, China
| | - Loulei Wang
- Medical Records Department, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hu Zhou
- General Committee Office, The People's Hospital of Yubei District of Chongqing City, Chongqing, China
| | - Yuan Yuan
- Medical Department, Women and Children's Hospital of Chongqing Medical University, Chongqing, China,*Correspondence: Yuan Yuan ✉
| | - Xiaoxin Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China,Xiaoxin Wu ✉
| |
Collapse
|
22
|
Mir MA, Bashir M, Ishfaq. Role of the CXCL8–CXCR1/2 Axis in Cancer and Inflammatory Diseases. CYTOKINE AND CHEMOKINE NETWORKS IN CANCER 2023:291-329. [DOI: 10.1007/978-981-99-4657-0_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
23
|
Speeckaert R, Belpaire A, Speeckaert MM, van Geel N. A meta-analysis of chemokines in vitiligo: Recruiting immune cells towards melanocytes. Front Immunol 2023; 14:1112811. [PMID: 36911664 PMCID: PMC9999440 DOI: 10.3389/fimmu.2023.1112811] [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: 11/30/2022] [Accepted: 02/16/2023] [Indexed: 03/14/2023] Open
Abstract
Chemokine research offers insightful information on the pathogenesis of cutaneous immune disorders, such as vitiligo. Compared to cytokines, the higher detectable levels of chemokines display promising potential as future disease biomarkers. Nonetheless, some published study results are contradictory, which can be attributed to patient characteristics and methodological differences. In this study, a meta-analysis was performed to compare chemokine expression in blood and skin samples from vitiligo patients versus healthy controls. Furthermore, the relationship between chemokine expression and disease activity was evaluated. Chemokine levels were investigated in 15 articles in the circulation and in 9 articles in vitiligo skin. Overall, some clear trends were observed. CXCR3 signaling by CXCL10 and CXCL9 has been confirmed by several reports, although CXCL10 showed more robust findings in blood samples. In this meta-analysis, CCL5, CXCL8, CXCL12, and CXCL16 levels were also significantly elevated. This indicates a complex immune pathway activation in vitiligo that overall supports a Th1-dominant response. Chemokines linked to the Th2 and Th17 pathways were less prevalent. Despite these findings, study protocols that examine a broader range of chemokines are encouraged, because current research is mostly focused on a small number of chemokines that were differentially expressed in previous studies.
Collapse
Affiliation(s)
| | - Arno Belpaire
- Department of Dermatology, Ghent University Hospital, Gent, Belgium
| | | | - Nanja van Geel
- Department of Dermatology, Ghent University Hospital, Gent, Belgium
| |
Collapse
|
24
|
Cui G, Liu H, Laugsand JB. Endothelial cells-directed angiogenesis in colorectal cancer: Interleukin as the mediator and pharmacological target. Int Immunopharmacol 2023; 114:109525. [PMID: 36508917 DOI: 10.1016/j.intimp.2022.109525] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/22/2022] [Accepted: 11/27/2022] [Indexed: 12/14/2022]
Abstract
Enhanced angiogenesis is a cancer hallmark and critical for colorectal cancer (CRC) invasion and metastasis. Upon exposure to proangiogenic factors, therefore, targeting tumor-associated proangiogenic factors/receptors hold great promise as a therapeutic modality to treat CRC, particularly metastatic CRC. Accumulating evidence from numerous studies suggests that tumor endothelial cells (ECs) are not only the target of proangiogenic factors, but also function as the cellular source of proangiogenic factors. Studies showed that ECs can produce different proangiogenic factors to participate in the regulation of angiogenesis process, in which ECs-derived interleukins (ILs) show a potential stimulatory effect on angiogenesis via either an direct action on their receptors expressed on progenitor of ECs or an indirect way through enhanced production of other proangiogenic factors. Although a great deal of attention is given to the effects of tumor-derived and immune cell-derived ILs, few studies describe the potential effects of vascular ECs-derived ILs on the tumor angiogenesis process. This review provides an updated summary of available information on proangiogenic ILs, such as IL-1, IL-6, IL-8, IL-17, IL-22, IL-33, IL-34, and IL-37, released by microvascular ECs as potential drivers of the tumor angiogenesis process and discusses their potential as a novel candidate for antiangiogenic target for the treatment of CRC patients.
Collapse
Affiliation(s)
- Guanglin Cui
- Research Group of Gastrointestinal Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; Faculty of Health Science, Nord University, Campus Levanger, Norway.
| | - Hanzhe Liu
- School of Stomatology, Wuhan University, Wuhan, China.
| | | |
Collapse
|
25
|
Zierfuss B, Buda A, Villoria-González A, Logist M, Fabjan J, Parzer P, Battin C, Vandersteene S, Dijkstra IME, Waidhofer-Söllner P, Grabmeier-Pfistershammer K, Steinberger P, Kemp S, Forss-Petter S, Berger J, Weinhofer I. Saturated very long-chain fatty acids regulate macrophage plasticity and invasiveness. J Neuroinflammation 2022; 19:305. [PMID: 36528616 PMCID: PMC9759912 DOI: 10.1186/s12974-022-02664-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 12/04/2022] [Indexed: 12/23/2022] Open
Abstract
Saturated very long-chain fatty acids (VLCFA, ≥ C22), enriched in brain myelin and innate immune cells, accumulate in X-linked adrenoleukodystrophy (X-ALD) due to inherited dysfunction of the peroxisomal VLCFA transporter ABCD1. In its severest form, X-ALD causes cerebral myelin destruction with infiltration of pro-inflammatory skewed monocytes/macrophages. How VLCFA levels relate to macrophage activation is unclear. Here, whole transcriptome sequencing of X-ALD macrophages indicated that VLCFAs prime human macrophage membranes for inflammation and increased expression of factors involved in chemotaxis and invasion. When added externally to mimic lipid release in demyelinating X-ALD lesions, VLCFAs did not activate toll-like receptors in primary macrophages. In contrast, VLCFAs provoked pro-inflammatory responses through scavenger receptor CD36-mediated uptake, cumulating in JNK signalling and expression of matrix-degrading enzymes and chemokine release. Following pro-inflammatory LPS activation, VLCFA levels increased also in healthy macrophages. With the onset of the resolution, VLCFAs were rapidly cleared in control macrophages by increased peroxisomal VLCFA degradation through liver-X-receptor mediated upregulation of ABCD1. ABCD1 deficiency impaired VLCFA homeostasis and prolonged pro-inflammatory gene expression upon LPS treatment. Our study uncovers a pivotal role for ABCD1, a protein linked to neuroinflammation, and associated peroxisomal VLCFA degradation in regulating macrophage plasticity.
Collapse
Affiliation(s)
- Bettina Zierfuss
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria
- Department of Neuroscience, Centre de Recherche du CHUM, Université de Montréal, Montréal, H2X 0A9, Canada
| | - Agnieszka Buda
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Andrea Villoria-González
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Maxime Logist
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria
- Department of Chronic Diseases and Metabolism, Translational Research in GastroIntestinal Disorders, KU Leuven, 3000, Leuven, Belgium
| | - Jure Fabjan
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Patricia Parzer
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Claire Battin
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Streggi Vandersteene
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Inge M E Dijkstra
- Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam Gastroenterology Endocrinology Metabolism, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Petra Waidhofer-Söllner
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Katharina Grabmeier-Pfistershammer
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Peter Steinberger
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Stephan Kemp
- Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam Gastroenterology Endocrinology Metabolism, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Sonja Forss-Petter
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Johannes Berger
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Isabelle Weinhofer
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria.
| |
Collapse
|
26
|
Xiang Q, Tao JS, Li JJ, Tian RB, Li XH. What is the role of Von Willebrand factor in chronic hepatitis B virus infection to hepatocellular carcinoma: a review article. Ther Adv Chronic Dis 2022; 13:20406223221125683. [PMID: 36407018 PMCID: PMC9669690 DOI: 10.1177/20406223221125683] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 08/25/2022] [Indexed: 11/12/2023] Open
Abstract
Von Willebrand factor (VWF) is a glycoprotein synthesized and secreted by vascular endothelial cells and megakaryocytes, found on plasma surface, endothelial cells, and α-granule of platelets. VWF can be interacted with collagen and platelet membrane glycoproteins GPIb and GPIb-IIa and play an important role in platelet adhesion and aggregation. Growing research evidence suggests that VWF also mediates the prevention or protesting of hepatocellular carcinoma (HCC) in chronic hepatitis B (CHB) patients from several clinical studies. While the mechanism of VWF in HCC protection or protest is still unclear, further study is required. This article aims to rationalize the role of VWF in the development of HCC, and the functional domain of VWF in cancer as well as cross-talking with platelets and miRNAs. This article also looks forward to the future development and challenges of VWF research.
Collapse
Affiliation(s)
- Qiong Xiang
- Medical Research Center, Institute of Medicine,
Jishou University, Jishou, China
| | - Jia-Sheng Tao
- Medical Research Center, Institute of Medicine,
Jishou University, Jishou, China
| | - Jing-Jing Li
- Medical Research Center, Institute of Medicine,
Jishou University, Jishou, China
| | - Rong-Bo Tian
- Medical Research Center, Institute of Medicine,
Jishou University, Jishou, China
| | - Xian-Hui Li
- Institute of Pharmaceutical Sciences, Jishou
University, 120 Ren min south road, Jishou 416000, China
| |
Collapse
|
27
|
Resistance to tyrosine kinase inhibitors promotes renal cancer progression through MCPIP1 tumor-suppressor downregulation and c-Met activation. Cell Death Dis 2022; 13:814. [PMID: 36138026 PMCID: PMC9500022 DOI: 10.1038/s41419-022-05251-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 02/08/2023]
Abstract
Tyrosine kinase inhibitors (TKIs) are the most commonly used targeted therapeutics in clear-cell renal cell carcinoma (ccRCC); however, drug resistance limits their utility and can lead to tumor "flare-up" and progression. In this study, we show that RCC resistance to sunitinib and sorafenib involves different mechanisms and leads to increased malignancy. Sunitinib decreased tumor growth and cell motility along with increased E-cadherin expression and secretion of the proangiogenic cytokines IL6 and IL8, which activated senescence in ccRCC cells and led to VE-cadherin phosphorylation, enhancing tumor angiogenesis. Sorafenib resistance increased the levels of mesenchymal markers and the secretion of MMP9, which cleaved VE-cadherin and disrupted endothelial cell integrity. Both sunitinib resistance and sorafenib resistance led to activation of the c-Met receptor IRAK1 and downregulation of the tumor suppressor MCPIP1, resulting in an increase in the metastasis of resistant cells, possibly due in part to enhanced vascularization of ccRCC. MCPIP1 overexpression partially overcame resistance to these drugs by decreasing micrometastasis and decreasing the expression of factors involved in tumorigenesis. In tumor samples from ccRCC patients, we observed a significant increase in the level of the c-Met receptor, IRAK1 and a decrease in MCPIP1 with respect to normal kidney tissue. Our results indicate separate novel mechanisms for sunitinib and sorafenib resistance, which both lead to MCPIP1 inhibition and ccRCC progression. The presented study suggests caution in the treatment of RCC with TKIs, which may lead to the unintended outcome of tumor progression.
Collapse
|
28
|
Targeting CXCR1 and CXCR2 receptors in cardiovascular diseases. Pharmacol Ther 2022; 237:108257. [PMID: 35908611 DOI: 10.1016/j.pharmthera.2022.108257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/21/2022] [Accepted: 07/25/2022] [Indexed: 11/22/2022]
|
29
|
Ciccone V, Terzuoli E, Ristori E, Filippelli A, Ziche M, Morbidelli L, Donnini S. ALDH1A1 overexpression in melanoma cells promotes tumor angiogenesis by activating the IL‑8/Notch signaling cascade. Int J Mol Med 2022; 50:99. [PMID: 35656893 PMCID: PMC9186295 DOI: 10.3892/ijmm.2022.5155] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 04/29/2022] [Indexed: 11/06/2022] Open
Abstract
ALDH1A1 is a cytosolic enzyme upregulated in tumor cells, involved in detoxifying cells from reactive aldehydes and in acquiring resistance to chemotherapeutic drugs. Its expression correlates with poor clinical outcomes in a number of cancers, including melanoma. The present study hypothesized that the increased ALDH1A1 expression and activity upregulated the release of proangiogenic factors from melanoma cells, which regulate angiogenic features in endothelial cells (ECs) through a rearrangement of the Notch pathway. In vivo, when subcutaneously implanted in immunodeficient mice, ALDH1A1 overexpressing melanoma cells displayed a higher microvessel density. In a 3D multicellular system, obtained co‑culturing melanoma cancer cells with stromal cells, including ECs, melanoma ALDH1A1 overexpression induced the recruitment of ECs into the core of the tumorspheres. By using a genes array, overexpression of ALDH1A1 in tumor cells also promoted modulation of Notch cascade gene expression in ECs, suggesting an interaction between tumor cells and ECs mediated by enrichment of angiogenic factors in the tumor microenvironment. To confirm this hypothesis, inactivation of ALDH1A1 by the pharmacological inhibitor CM037 significantly affected the release of angiogenic factors, including IL‑8, from melanoma cells. High levels of ALDH1A1, through the retinoic acid pathway, regulated the activation of NF‑kB‑p65 and IL‑8. Further, in a 2D co‑culture system, the addition of an IL‑8 neutralizing antibody to ECs co‑cultured with melanoma cells forced to express ALDH1A1 dampened endothelial angiogenic features, both at the molecular (in terms of gene and protein expression of mediators of the Notch pathway) and at the functional level (proliferation, scratch assay, tube formation and permeability). In conclusion, these findings demonstrated the existence of a link between melanoma ALDH1A1 expression and EC Notch signaling modification that results in a pro‑angiogenic phenotype. Based on the crucial role of ALDH1A1 in melanoma control of the tumor microenvironment, the enzyme seems a promising target for the development of novel drugs able to interrupt the cross‑talk between cancer (stem) cells and endothelial cells.
Collapse
Affiliation(s)
- Valerio Ciccone
- Department of Life Sciences, University of Siena, Siena I‑53100, Italy
| | - Erika Terzuoli
- Department of Life Sciences, University of Siena, Siena I‑53100, Italy
| | - Emma Ristori
- Department of Life Sciences, University of Siena, Siena I‑53100, Italy
| | | | - Marina Ziche
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena I‑53100, Italy
| | - Lucia Morbidelli
- Department of Life Sciences, University of Siena, Siena I‑53100, Italy
| | - Sandra Donnini
- Department of Life Sciences, University of Siena, Siena I‑53100, Italy
| |
Collapse
|
30
|
Yao X, He Z, Qin C, Zhang P, Sui C, Deng X, Fang Y, Li G, Shi J. Inhibition of PFKFB3 in HER2-positive gastric cancer improves sensitivity to trastuzumab by inducing tumour vessel normalisation. Br J Cancer 2022; 127:811-823. [PMID: 35637411 DOI: 10.1038/s41416-022-01834-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 04/14/2022] [Accepted: 04/26/2022] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Multiple mechanisms have been proposed that lead to reduced effectiveness of trastuzumab in HER2-positive gastric cancer (GC), yet resistance to trastuzumab remains a challenge in clinics. METHODS We established trastuzumab-resistant cells and patient-derived xenografts models to measure metabolic levels and vascular density and shape. The HER2-positive GC patient samples were used to determine clinical significance. We also measured protein expression and phosphorylation modifications to determine those alterations related to resistance. In vivo studies combining inhibitor of PFKFB3 with trastuzumab corroborated the in vitro findings. RESULTS The 6-phosphofructo-2-kinase (PFKFB3)-mediated trastuzumab resistance pathways in HER2-positive GC by activating the glycolytic pathway. We also found vessels are chaotic and destabilised in the tumour during the trastuzumab resistance process. Inhibition of PFKFB3 significantly diminished tumour proliferation and promoted vessel normalisation in the patient-derived xenograft model. Mechanistically, PFKFB3 promoted the secretion of CXCL8 into the tumour microenvironment, and phosphorylated Ser1151 of ERBB2, enhancing the transcription of CXCL8 by activating the PI3K/AKT/NFκB p65 pathway. CONCLUSIONS Our current findings discover that PFKFB3 inhibitors might be effective tools to overcome adjuvant therapy resistance in HER2-positive GC and reshaping the microenvironment by normalising tumour vessels is a novel strategy to overcome trastuzumab resistance.
Collapse
Affiliation(s)
- Xingxing Yao
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, 510515, Guangzhou, Guangdong, China
| | - Zhanke He
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, 510515, Guangzhou, Guangdong, China
| | - Caolitao Qin
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases & Department of Radiation Oncology, The Sixth Affiliated Hospital of Sun Yat-sen University, 510655, Guangzhou, China
| | - Penghao Zhang
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, 510515, Guangzhou, Guangdong, China
| | - Chuyang Sui
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, 510515, Guangzhou, Guangdong, China
| | - Xiangqian Deng
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, 510515, Guangzhou, Guangdong, China
| | - Yuxin Fang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Guoxin Li
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, 510515, Guangzhou, Guangdong, China.
| | - Jiaolong Shi
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, 510515, Guangzhou, Guangdong, China.
| |
Collapse
|
31
|
Lu X, Wang Z, Ye D, Feng Y, Liu M, Xu Y, Wang M, Zhang J, Liu J, Zhao M, Xu S, Ye J, Wan J. The Role of CXC Chemokines in Cardiovascular Diseases. Front Pharmacol 2022; 12:765768. [PMID: 35668739 PMCID: PMC9163960 DOI: 10.3389/fphar.2021.765768] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 12/08/2021] [Indexed: 01/07/2023] Open
Abstract
Cardiovascular disease (CVD) is a class of diseases with high disability and mortality rates. In the elderly population, the incidence of cardiovascular disease is increasing annually. Between 1990 and 2016, the age-standardised prevalence of CVD in China significantly increased by 14.7%, and the number of cardiovascular disease deaths increased from 2.51 million to 3.97 million. Much research has indicated that cardiovascular disease is closely related to inflammation, immunity, injury and repair. Chemokines, which induce directed chemotaxis of reactive cells, are divided into four subfamilies: CXC, CC, CX3C, and XC. As cytokines, CXC chemokines are similarly involved in inflammation, immunity, injury, and repair and play a role in many cardiovascular diseases, such as atherosclerosis, myocardial infarction, cardiac ischaemia-reperfusion injury, hypertension, aortic aneurysm, cardiac fibrosis, postcardiac rejection, and atrial fibrillation. Here, we explored the relationship between the chemokine CXC subset and cardiovascular disease and its mechanism of action with the goal of further understanding the onset of cardiovascular disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Jing Ye
- *Correspondence: Jing Ye, ; Jun Wan,
| | - Jun Wan
- *Correspondence: Jing Ye, ; Jun Wan,
| |
Collapse
|
32
|
Mateluna C, Torres P, Rodriguez-Peña M, Silva P, Matthies DJ, Criollo A, Bikker FJ, Bolscher JGM, Wilson CAM, Zapata-Torres G, Torres VA. Identification of VEGFR2 as the Histatin-1 receptor in endothelial cells. Biochem Pharmacol 2022; 201:115079. [PMID: 35551916 DOI: 10.1016/j.bcp.2022.115079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 04/18/2022] [Accepted: 05/02/2022] [Indexed: 11/02/2022]
Abstract
Histatin-1 is a salivary peptide with antimicrobial and wound healing promoting activities, which was previously shown to stimulate angiogenesis in vitro and in vivo via inducing endothelial cell migration. The mechanisms underlying the proangiogenic effects of Histatin-1 remain poorly understood and specifically, the endothelial receptor for this peptide, is unknown. Based on the similarities between Histatin-1-dependent responses and those induced by the prototypical angiogenic receptor, vascular endothelial growth factor receptor 2 (VEGFR2), we hypothesized that VEGFR2 is the Histatin-1 receptor in endothelial cells. First, we observed that VEGFR2 is necessary for Histatin-1-induced endothelial cell migration, as shown by both pharmacological inhibition studies and siRNA-mediated ablation of VEGFR2. Moreover, Histatin-1 co-immunoprecipitated and co-localized with VEGFR2, associating spatial proximity between these proteins with receptor activation. Indeed, pulldown assays with pure, tagged and non-tagged proteins showed that Histatin-1 and VEGFR2 directly interact in vitro. Optical tweezers experiments permitted estimating kinetic parameters and rupture forces, indicating that the Histatin-1-VEGFR2 interaction is transient, but specific and direct. Sequence alignment and molecular modeling identified residues Phe26, Tyr30 and Tyr34 within the C-terminal domain of Histatin-1 as relevant for VEGFR2 binding and activation. This was corroborated by mutation and molecular dynamics analyses, as well as in direct binding assays. Importantly, these residues were required for Histatin-1 to induce endothelial cell migration and angiogenesis in vitro. Taken together, our findings reveal that VEGFR2 is the endothelial cell receptor of Histatin-1 and provide insights to the mechanism by which this peptide promotes endothelial cell migration and angiogenesis.
Collapse
Affiliation(s)
- Carlos Mateluna
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile; Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santiago, Chile
| | - Pedro Torres
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile; Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santiago, Chile
| | - Marcelo Rodriguez-Peña
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile; Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santiago, Chile
| | - Patricio Silva
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile; Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santiago, Chile
| | - Douglas J Matthies
- Molecular Graphics Suite, Department of Inorganic and Analytical Chemistry, Faculty of Chemical and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile
| | - Alfredo Criollo
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile; Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santiago, Chile
| | - Floris J Bikker
- Department of Oral Biochemistry, Academic Centre for Dentistry Amsterdam, VU University & University of Amsterdam, Netherlands
| | - Jan G M Bolscher
- Department of Oral Biochemistry, Academic Centre for Dentistry Amsterdam, VU University & University of Amsterdam, Netherlands
| | - Christian A M Wilson
- Department of Biochemistry and Molecular Biology, Faculty of Chemical and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile
| | - Gerald Zapata-Torres
- Molecular Graphics Suite, Department of Inorganic and Analytical Chemistry, Faculty of Chemical and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile
| | - Vicente A Torres
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile; Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santiago, Chile; Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
33
|
Xiong X, Liao X, Qiu S, Xu H, Zhang S, Wang S, Ai J, Yang L. CXCL8 in Tumor Biology and Its Implications for Clinical Translation. Front Mol Biosci 2022; 9:723846. [PMID: 35372515 PMCID: PMC8965068 DOI: 10.3389/fmolb.2022.723846] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 02/21/2022] [Indexed: 12/13/2022] Open
Abstract
The chemokine CXCL8 has been found to play an important role in tumor progression in recent years. CXCL8 activates multiple intracellular signaling pathways by binding to its receptors (CXCR1/2), and plays dual pro-tumorigenic roles in the tumor microenvironment (TME) including directly promoting tumor survival and affecting components of TME to indirectly facilitate tumor progression, which include facilitating tumor cell proliferation and epithelial-to-mesenchymal transition (EMT), pro-angiogenesis, and inhibit anti-tumor immunity. More recently, clinical trials indicate that CXCL8 can act as an independently predictive biomarker in patients receiving immune checkpoint inhibitions (ICIs) therapy. Preclinical studies also suggest that combined CXCL8 blockade and ICIs therapy can enhance the anti-tumor efficacy, and several clinical trials are being conducted to evaluate this therapy modality.
Collapse
Affiliation(s)
- Xingyu Xiong
- Department of Urology, National Clinical Research Center for Geriatrics, Institute of Urology, West China Hospital of Sichuan University, Chengdu, China
| | - Xinyang Liao
- Department of Urology, National Clinical Research Center for Geriatrics, Institute of Urology, West China Hospital of Sichuan University, Chengdu, China
| | - Shi Qiu
- Department of Urology, National Clinical Research Center for Geriatrics, Institute of Urology, West China Hospital of Sichuan University, Chengdu, China
- Center of Biomedical Big Data, West China Hospital, Sichuan University, Chengdu, China
| | - Hang Xu
- Department of Urology, National Clinical Research Center for Geriatrics, Institute of Urology, West China Hospital of Sichuan University, Chengdu, China
| | - Shiyu Zhang
- Department of Urology, National Clinical Research Center for Geriatrics, Institute of Urology, West China Hospital of Sichuan University, Chengdu, China
| | - Sheng Wang
- Department of Urology, National Clinical Research Center for Geriatrics, Institute of Urology, West China Hospital of Sichuan University, Chengdu, China
| | - Jianzhong Ai
- Department of Urology, National Clinical Research Center for Geriatrics, Institute of Urology, West China Hospital of Sichuan University, Chengdu, China
- *Correspondence: Jianzhong Ai, ; Lu Yang,
| | - Lu Yang
- Department of Urology, National Clinical Research Center for Geriatrics, Institute of Urology, West China Hospital of Sichuan University, Chengdu, China
- *Correspondence: Jianzhong Ai, ; Lu Yang,
| |
Collapse
|
34
|
Silva ALM, Silva ECO, Botelho RM, Tenorio LPG, Marques ALX, Rodrigues IBAC, Almeida LIM, Sousa AKA, Pires KSN, Tanabe ISB, Allard MJ, Sébire G, Souza ST, Fonseca EJS, Borbely KSC, Borbely AU. Uvaol Prevents Group B Streptococcus-Induced Trophoblast Cells Inflammation and Possible Endothelial Dysfunction. Front Physiol 2021; 12:766382. [PMID: 34925062 PMCID: PMC8678414 DOI: 10.3389/fphys.2021.766382] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 11/02/2021] [Indexed: 12/25/2022] Open
Abstract
Group B Streptococcus (GBS) infection during pregnancy is involved in maternal sepsis, chorioamnionitis, prematurity, fetal infection, neonatal sepsis, and neurodevelopmental alterations. The GBS-induced chorioamnionitis leads to a plethora of immune and trophoblast cells alterations that could influence endothelial cells to respond differently to angiogenic mediators and alter placental vascular structure and function in pregnant women. In this context, preventive measures are needed to reduce such dysfunctions. As such, we evaluated the effects of a non-lethal exposure to inactivated GBS on trophoblast cells and chorionic villi explants, and if the treatment with uvaol would mitigate these effects. The concentration of 106 CFU of GBS was chosen since it was unable to reduce the HTR-8/SVneo cell line nor term chorionic villi explant viability. Raman spectroscopy of trophoblast cells showed significant alterations in their biochemical signature, mostly reverted by uvaol. GBS exposure increased HTR-8/SVneo cells IL-1β and IFN-γ production, phagocytosis, oxidative stress, and decreased trophoblast cell migration. The Ea.hy926 endothelial cell line produced angiopoietin-2, CXCL-8, EGF, FGF-b, IL-6, PlGF, sPECAM-1, and VEGF in culture. When co-cultured in invasion assay with HTR-8/SVneo trophoblast cells, the co-culture had increased production of angiopoietin-2, CXCL-8, FGF-b, and VEGF, while reduced sPECAM-1 and IL-6. GBS exposure led to increased CXCL-8 and IL-6 production, both prevented by uvaol. Chorionic villi explants followed the same patterns of production when exposed to GBS and response to uvaol treatment as well. These findings demonstrate that, even a non-lethal concentration of GBS causes placental inflammation and oxidative stress, reduces trophoblast invasion of endothelial cells, and increases CXCL-8 and IL-6, key factors that participate in vascular dysregulation observed in several diseases. Furthermore, uvaol treatment prevented most of the GBS-provoked changes. Hence, uvaol could prevent the harmful effects of GBS infection for both the mother and the fetus.
Collapse
Affiliation(s)
- Ana Lucia Mendes Silva
- Cell Biology Laboratory, Institute of Health and Biological Sciences, Federal University of Alagoas, Maceio, Brazil
| | | | - Rayane Martins Botelho
- Cell Biology Laboratory, Institute of Health and Biological Sciences, Federal University of Alagoas, Maceio, Brazil
| | | | - Aldilane Lays Xavier Marques
- Cell Biology Laboratory, Institute of Health and Biological Sciences, Federal University of Alagoas, Maceio, Brazil
| | | | | | | | - Keyla Silva Nobre Pires
- Cell Biology Laboratory, Institute of Health and Biological Sciences, Federal University of Alagoas, Maceio, Brazil
| | | | | | - Guillaume Sébire
- Department of Pediatrics, McGill University, Montreal, QC, Canada.,Department of Pediatrics, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Samuel Teixeira Souza
- Optics and Nanoscopy Group, Physics Institute, Federal University of Alagoas, Maceio, Brazil
| | | | - Karen Steponavicius Cruz Borbely
- Cell Biology Laboratory, Institute of Health and Biological Sciences, Federal University of Alagoas, Maceio, Brazil.,Faculty of Nutrition, Federal University of Alagoas, Maceio, Brazil
| | - Alexandre Urban Borbely
- Cell Biology Laboratory, Institute of Health and Biological Sciences, Federal University of Alagoas, Maceio, Brazil
| |
Collapse
|
35
|
Clinical Update of Severe Fever with Thrombocytopenia Syndrome. Viruses 2021; 13:v13071213. [PMID: 34201811 PMCID: PMC8310018 DOI: 10.3390/v13071213] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 06/18/2021] [Accepted: 06/18/2021] [Indexed: 12/17/2022] Open
Abstract
Severe fever with thrombocytopenia syndrome (SFTS) is an acute febrile illness characterized by fever, leukopenia, thrombocytopenia, and gastrointestinal symptoms such as diarrhea, nausea, and vomiting resulting from infection with the SFTS virus (SFTSV). The SFTSV is transmitted to humans by tick bites, primarily from Haemaphysalis longicornis, Amblyomma testudinarium, Ixodes nipponensis, and Rhipicephalus microplus. Human-to-human transmission has also been reported. Since the first report of an SFTS patient in China, the number of patients has also been increasing. The mortality rate of patients with SFTS remains high because the disease can quickly lead to death through multiple organ failure. In particular, an average fatality rate of approximately 20% has been reported for SFTS patients, and no treatment strategy has been established. Therefore, effective antiviral agents and vaccines are required. Here, we aim to review the epidemiology, clinical manifestations, laboratory diagnosis, and various specific treatments (i.e., antiviral agents, steroids, intravenous immunoglobulin, and plasma exchange) that have been tested to help to cope with the disease.
Collapse
|
36
|
Tomita T, Kato M, Hiratsuka S. Regulation of vascular permeability in cancer metastasis. Cancer Sci 2021; 112:2966-2974. [PMID: 33966313 PMCID: PMC8353911 DOI: 10.1111/cas.14942] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/27/2021] [Accepted: 05/03/2021] [Indexed: 12/14/2022] Open
Abstract
Enhancement of vascular permeability is indispensable for cancer metastasis. Weakened endothelial barrier function enhances vascular permeability. Circulating tumor cells moving in the microvasculature tend to invade into stromal tissue at the location where vascular permeability is enhanced. Many basic studies have identified permeability factors by using gene‐modified animals and cells. These factors directly/indirectly interact with endothelial cells. Here, we review vascular permeability factors and their molecular mechanisms. Interactions between tumor cells and endothelial cells are also discussed in the process of extravasation, one of the most critical steps in tumor metastasis. In some cases, primary tumors can manipulate permeability in a remote organ by secreting permeability factors. In addition, the importance of glycocalyx, which covers the endothelial cell surface, in controlling vascular permeability and tumor metastasis is also described. Furthermore, analysis of the hyperpermeable region found in a mouse model study is introduced. It clearly showed that tumor‐bearing mouse lungs had a hyperpermeable region due to the influence of a remote primary tumor, and fibrinogen deposition was observed in that region. Given that fibrinogen was reported to be a permeability factor and a key regulator of inflammation, eliminating fibrinogen deposition may prevent future metastasis.
Collapse
Affiliation(s)
- Takeshi Tomita
- Department of Biochemistry and Molecular Biology, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University School of Medicine, Matsumoto, Japan
| | - Masayoshi Kato
- Department of Biochemistry and Molecular Biology, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University School of Medicine, Matsumoto, Japan
| | - Sachie Hiratsuka
- Department of Biochemistry and Molecular Biology, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University School of Medicine, Matsumoto, Japan
| |
Collapse
|
37
|
Sharma D, Kamthania M. A new emerging pandemic of severe fever with thrombocytopenia syndrome (SFTS). Virusdisease 2021; 32:220-227. [PMID: 33942022 PMCID: PMC8082055 DOI: 10.1007/s13337-021-00656-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 01/18/2021] [Indexed: 02/07/2023] Open
Abstract
The aim of this study is to make aware every one of the deadliest diseases named severe fever with thrombocytopenia syndrome (SFTS). It has become the worldwide pandemic in recent few years. It is a kind of haemorrhagic fever, caused by SFTS virus (SFTSV), a novel phlebovirus of family Bunyaviridae. This syndrome is also a tick-borne zoonosis that means the virus transmitted from tick bite (having virus) into human body, i.e. infection spread from animals to humans and also transmitted from human to human. Epidemiological data of SFTS was collected to know the nature/symptoms of SFTSV. First case of this disease has been reported in China, followed by Japan, South korea, Taiwan, USA and many other countries. Vertebrates are the host of this disease and tick functions as a vector, where the virus can undergo brisk changes using gene mutation, homologous recombination and reassortments. The major symptoms of hemorrhagic fever are fever, thrombocytopenia, leucopenia and gastrointestinal abnormalities. Sometimes in very severe cases, full body organ failure may also take place and average death rate in humans is nearly 10 %. Old aged peoples are more prone to SFTSV infection. Apart from the fact of increasing SFTSV related health problems to humans, the pathogenesis of SFTS virus in human is not entirely understood and no treatment to this virus is still available. The simplest way to protect our self from this infection is to refrain from tick bite. Therefore, this disease has evolved to produce serious health issues to humans in various countries of world including china. This review discussing about causative agent, epidemiology, pathogenesis, diagnosis and treatment of SFTS. In order to control the spread of SFTSV, we have to stop the viral transmission or to protect the easily vulnerable population from tick bites, avoiding direct contact of infectious and also to use personal protective devices for SFTS patients. So, the weather conditions, mode of transmission and creation of new therapeutics like vaccines and drugs are the main areas of forthcoming research.
Collapse
Affiliation(s)
- Divya Sharma
- Department of Life Sciences, IAMR College, Ghaziabad, Uttar Pradesh India
| | - Mohit Kamthania
- Department of Life Sciences, IAMR College, Ghaziabad, Uttar Pradesh India
| |
Collapse
|
38
|
The CXCL2/IL8/CXCR2 Pathway Is Relevant for Brain Tumor Malignancy and Endothelial Cell Function. Int J Mol Sci 2021; 22:ijms22052634. [PMID: 33807899 PMCID: PMC7961945 DOI: 10.3390/ijms22052634] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 12/30/2022] Open
Abstract
We aimed to evaluate the angiogenic capacity of CXCL2 and IL8 affecting human endothelial cells to clarify their potential role in glioblastoma (GBM) angiogenesis. Human GBM samples and controls were stained for proangiogenic factors. Survival curves and molecule correlations were obtained from the TCGA (The Cancer Genome Atlas) database. Moreover, proliferative, migratory and angiogenic activity of peripheral (HUVEC) and brain specific (HBMEC) primary human endothelial cells were investigated including blockage of CXCR2 signaling with SB225502. Gene expression analyses of angiogenic molecules from endothelial cells were performed. Overexpression of VEGF and CXCL2 was observed in GBM patients and associated with a survival disadvantage. Molecules of the VEGF pathway correlated but no relation for CXCR1/2 and CXCL2/IL8 was found. Interestingly, receptors of endothelial cells were not induced by addition of proangiogenic factors in vitro. Proliferation and migration of HUVEC were increased by VEGF, CXCL2 as well as IL8. Their sprouting was enhanced through VEGF and CXCL2, while IL8 showed no effect. In contrast, brain endothelial cells reacted to all proangiogenic molecules. Additionally, treatment with a CXCR2 antagonist led to reduced chemokinesis and sprouting of endothelial cells. We demonstrate the impact of CXCR2 signaling on endothelial cells supporting an impact of this pathway in angiogenesis of glioblastoma.
Collapse
|
39
|
Chen YC, Dinavahi SS, Feng Q, Gowda R, Ramisetti S, Xia X, LaPenna KB, Chirasani VR, Cho SH, Hafenstein SL, Battu MB, Berg A, Sharma AK, Kirchhausen T, Dokholyan NV, Amin S, He P, Robertson GP. Activating Sphingosine-1-phospahte signaling in endothelial cells increases myosin light chain phosphorylation to decrease endothelial permeability thereby inhibiting cancer metastasis. Cancer Lett 2021; 506:107-119. [PMID: 33600895 DOI: 10.1016/j.canlet.2021.01.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 12/22/2020] [Accepted: 01/04/2021] [Indexed: 12/23/2022]
Abstract
Targeting the metastatic process to prevent disease dissemination in cancer remains challenging. One step in the metastatic cascade involves cancer cells transiting through the vascular endothelium after inflammation has increased the permeability of this cellular layer. Reducing inflammation-mediated gaps in the vascular endothelium could potentially be used to retard metastasis. This study describes the development of a novel ASR396-containing nanoparticle designed to activate the Sphingosine-1-Phosphate Receptor 1 (S1PR1) in order to tighten the junctions between the endothelial cells lining the vascular endothelium thereby inhibiting metastasis. ASR396 was derived from the S1PR1 agonist SEW2871 through chemical modification enabling the new compound to be loaded into a nanoliposome. ASR396 retained S1PR1 binding activity and the nanoliposomal formulation (nanoASR396) made it systemically bioavailable upon intravenous injection. Studies conducted in microvessels demonstrated that nanoASR396 significantly attenuated inflammatory mediator-induced permeability increase through the S1PR1 activation. Similarly, nanoASR396 inhibited gap formation mediated by inflammatory agents on an endothelial cell monolayer by decreasing levels of phosphorylated myosin light chain protein thereby inhibiting cellular contractility. In animal models, nanoASR396 inhibited lung metastasis by up to 80%, indicating its potential for retarding melanoma metastasis. Thus, a novel bioavailable nanoparticle-based S1PR1 agonist has been developed to negate the effects of inflammatory mediators on the vascular endothelium in order to reduce the metastatic dissemination of cancer cells.
Collapse
Affiliation(s)
- Yu-Chi Chen
- Departments of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Saketh S Dinavahi
- Departments of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Qilong Feng
- Departments of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Raghavendra Gowda
- Departments of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Srinivasa Ramisetti
- Departments of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Xinghai Xia
- Departments of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Kyle B LaPenna
- Departments of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Venkat R Chirasani
- Departments of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Sung Hyun Cho
- The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA
| | - Susan L Hafenstein
- Departments of Medicine, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA; Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, 16802, USA
| | | | - Arthur Berg
- Departments of Public Health Sciences, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Arun K Sharma
- Departments of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Tom Kirchhausen
- Departments of Cell Biology, Harvard Medical School and Program in Cellular and Molecular Medicine at Boston Children's Hospital, MA, 02115, USA
| | - Nikolay V Dokholyan
- Departments of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA; Departments of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Shantu Amin
- Departments of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
| | - Pingnian He
- Departments of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
| | - Gavin P Robertson
- Departments of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA; Departments of Departments of Pathology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA; Departments of Dermatology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA; Departments of Surgery, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA; The Foreman Foundation for Melanoma Research, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA; The Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA; The Melanoma Therapeutics Program, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
| |
Collapse
|
40
|
Han L, Korangath P, Nguyen NK, Diehl A, Cho S, Teo WW, Cope L, Gessler M, Romer L, Sukumar S. HEYL Regulates Neoangiogenesis Through Overexpression in Both Breast Tumor Epithelium and Endothelium. Front Oncol 2021; 10:581459. [PMID: 33520697 PMCID: PMC7845423 DOI: 10.3389/fonc.2020.581459] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 11/30/2020] [Indexed: 12/20/2022] Open
Abstract
Blocking tumor angiogenesis is an appealing therapeutic strategy, but to date, success has been elusive. We previously identified HEYL, a downstream target of Notch signaling, as an overexpressed gene in both breast cancer cells and as a tumor endothelial marker, suggesting that HEYL overexpression in both compartments may contribute to neoangiogenesis. Carcinomas arising in double transgenic Her2-neu/HeyL mice showed higher tumor vessel density and significantly faster growth than tumors in parental Her2/neu mice. Providing mechanistic insight, microarray-based mRNA profiling of HS578T-tet-off-HEYL human breast cancer cells revealed upregulation of several angiogenic factors including CXCL1/2/3 upon HEYL expression, which was validated by RT-qPCR and protein array analysis. Upregulation of the cytokines CXCL1/2/3 occurred through direct binding of HEYL to their promoter sequences. We found that vessel growth and migration of human vascular endothelial cells (HUVECs) was promoted by conditioned medium from HS578T-tet-off-HEYL carcinoma cells, but was blocked by neutralizing antibodies against CXCL1/2/3. Supporting these findings, suppressing HEYL expression using shRNA in MDA-MB-231 cells significantly reduced tumor growth. In addition, suppressing the action of proangiogenic cytokines induced by HEYL using a small molecule inhibitor of the CXCl1/2/3 receptor, CXCR2, in combination with the anti-VEGF monoclonal antibody, bevacizumab, significantly reduced tumor growth of MDA-MB-231 xenografts. Thus, HEYL expression in tumor epithelium has a profound effect on the vascular microenvironment in promoting neoangiogenesis. Furthermore, we show that lack of HEYL expression in endothelial cells leads to defects in neoangiogenesis, both under normal physiological conditions and in cancer. Thus, HeyL-/- mice showed impaired vessel outgrowth in the neonatal retina, while the growth of mammary tumor cells E0771 was retarded in syngeneic HeyL-/- mice compared to wild type C57/Bl6 mice. Blocking HEYL's angiogenesis-promoting function in both tumor cells and tumor-associated endothelium may enhance efficacy of therapy targeting the tumor vasculature in breast cancer.
Collapse
Affiliation(s)
- Liangfeng Han
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Preethi Korangath
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Nguyen K Nguyen
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Adam Diehl
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Soonweng Cho
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Wei Wen Teo
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Leslie Cope
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Manfred Gessler
- Developmental Biochemistry, Comprehensive Cancer Center Mainfraken and Theodor-Boveri-Institute/Biocenter, University of Wurzburg, Wurzburg, Germany
| | - Lewis Romer
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,The Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Saraswati Sukumar
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
41
|
Giannetta E, La Salvia A, Rizza L, Muscogiuri G, Campione S, Pozza C, Colao AALI, Faggiano A. Are Markers of Systemic Inflammatory Response Useful in the Management of Patients With Neuroendocrine Neoplasms? Front Endocrinol (Lausanne) 2021; 12:672499. [PMID: 34367064 PMCID: PMC8339959 DOI: 10.3389/fendo.2021.672499] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/03/2021] [Indexed: 11/13/2022] Open
Abstract
Given the increasing incidence of neuroendocrine neoplasms (NENs) over the past few decades, a more comprehensive knowledge of their pathophysiological bases and the identification of innovative NEN biomarkers represents an urgent unmet need. There is still little advance in the early diagnosis and management of these tumors, due to the lack of sensible and specific markers with prognostic value and ability to early detect the response to treatment. Chronic systemic inflammation is a predisposing factor for multiple cancer hallmarks, as cancer proliferation, progression and immune-evading. Therefore, the relevance of inflammatory biomarkers has been identified as critical in several types of tumours, including NENs. A bidirectional relationship between chronic inflammation and development of NENs has been reported. Neuroendocrine cells can be over-stimulated by chronic inflammation, leading to hyperplasia and neoplastic transformation. As the modulation of inflammatory response represents a therapeutic target, inflammatory markers could represent a promising new key tool to be applied in the diagnosis, the prediction of response to treatment and also as prognostic biomarkers in NENs field. The present review provides an overview of the pre-clinical and clinical data relating the potentially usefulness of circulating inflammatory markers: neutrophil-lymphocyte ratio (NLR), platelet-lymphocyte ratio (PLR), cytokines and tissue inflammatory markers (PD-1/PD-L1), in the management of NENs. (1) NLR and PLR have both demonstrated to be promising and simple to acquire biomarkers in patients with advanced cancer, including NEN. To date, in the context of NENs, the prognostic role of NLR and PLR has been confirmed in 15 and 4 studies, respectively. However, the threshold value, both for NLR and PLR, still remains not defined. (2) Cytokines seem to play a central role in NENs tumorigenesis. In particular, IL-8 levels seems to be a good predictive marker of response to anti-angiogenic treatments. (3) PD-1 and PD-L1 expression on tumour cells and on TILs, have demonstrated to be promising predictive and prognostic biomarkers in NENs. Unfortunately, these two markers have not been validated so far and further studies are needed to establish their indications and utility.
Collapse
Affiliation(s)
- Elisa Giannetta
- Department of Experimental Medicine, “Sapienza” University of Rome, Rome, Italy
- *Correspondence: Elisa Giannetta,
| | - Anna La Salvia
- Department of Oncology, University Hospital 12 de Octubre, Madrid, Spain
| | - Laura Rizza
- Endocrinology Unit, Department of Oncology and Medical Specialities, AO San Camillo-Forlanini, Rome, Italy
| | - Giovanna Muscogiuri
- Endocrinology Unit Department of Clinical Medicine and Surgery, University Federico II School of Medicine, Naples, Italy
| | - Severo Campione
- A. Cardarelli Hospital, Naples Department of Advanced Diagnostic-Therapeutic Technologies and Health Services Section of Anatomic Pathology, Naples, Italy
| | - Carlotta Pozza
- Department of Experimental Medicine, “Sapienza” University of Rome, Rome, Italy
| | | | - Antongiulio Faggiano
- Department of Clinical and Molecular Medicine, Endocrine-Metabolic Unit, Sant’Andrea University Hospital “Sapienza” University of Rome, Rome, Italy
| |
Collapse
|
42
|
Torres MA, Gualtero DF, Lafaurie GI, Fontanilla MR. Aggregatibacter actinomycetemcomitans
induces a proatherosclerotic response in human endothelial cells in a three‐dimensional collagen scaffold model. Mol Oral Microbiol 2020. [DOI: 10.1111/omi.12326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Maria A. Torres
- Tissue Engineering Group Department of Pharmacy Universidad Nacional de Colombia Bogotá Colombia
- Biotechnology Laboratory, Basic Oral Research Unit (UIBO) School of Odontology Universidad El Bosque Bogotá Colombia
| | - Diego F. Gualtero
- Tissue Engineering Group Department of Pharmacy Universidad Nacional de Colombia Bogotá Colombia
- Biotechnology Laboratory, Basic Oral Research Unit (UIBO) School of Odontology Universidad El Bosque Bogotá Colombia
| | - Gloria I. Lafaurie
- Biotechnology Laboratory, Basic Oral Research Unit (UIBO) School of Odontology Universidad El Bosque Bogotá Colombia
| | - Marta R. Fontanilla
- Tissue Engineering Group Department of Pharmacy Universidad Nacional de Colombia Bogotá Colombia
| |
Collapse
|
43
|
The Role of Macrophages in Vascular Repair and Regeneration after Ischemic Injury. Int J Mol Sci 2020; 21:ijms21176328. [PMID: 32878297 PMCID: PMC7503238 DOI: 10.3390/ijms21176328] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/25/2020] [Accepted: 08/26/2020] [Indexed: 12/11/2022] Open
Abstract
Macrophage is one of the important players in immune response which perform many different functions during tissue injury, repair, and regeneration. Studies using animal models of cardiovascular diseases have provided a clear picture describing the effect of macrophages and their phenotype during injury and regeneration of various vascular beds. Many data have been generated to demonstrate that macrophages secrete many important factors including cytokines and growth factors to regulate angiogenesis and arteriogenesis, acting directly or indirectly on the vascular cells. Different subsets of macrophages may participate at different stages of vascular repair. Recent findings also suggest a direct interaction between macrophages and other cell types during the generation and repair of vasculature. In this short review, we focused our discussion on how macrophages adapt to the surrounding microenvironment and their potential interaction with other cells, in the context of vascular repair supported by evidences mostly from studies using hindlimb ischemia as a model for studying post-ischemic vascular repair.
Collapse
|
44
|
Zhou LB, Zhou YQ, Zhang XY. Blocking VEGF signaling augments interleukin-8 secretion via MEK/ERK/1/2 axis in human retinal pigment epithelial cells. Int J Ophthalmol 2020; 13:1039-1045. [PMID: 32685389 PMCID: PMC7321944 DOI: 10.18240/ijo.2020.07.04] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 04/23/2020] [Indexed: 12/13/2022] Open
Abstract
AIM To identify proangiogenic factors engaged in neovascular age-related macular degeneration (AMD) except vascular endothelial growth factor (VEGF) from human retinal pigment epithelial (hRPE) cells and investigate the underlying mechanisms. METHODS VEGF receptor 2 (VEGFR2) in ARPE-19 cells was depleted by siRNA transfection or overexpressed through adenovirus infection. The mRNA and the protein levels of interleukin-8 (IL-8) in ARPE-19 cells were measured by quantitative real-time polymerase chain reaction and enzyme-linked immunosorbent assay respectively. The protein levels of AKT, p-AKT, MEK, p-MEK, ERK1/2, p-ERK1/2, JNK, p-JNK, p38 and p-p38 were detected by Western blotting. A selective chemical inhibitor, LY3214996, was employed to inhibit phosphorylation of ERK1/2. Cell viability was determined by MTT assay. RESULTS Knockdown of VEGFR2 in ARPE-19 cells robustly augmented IL-8 production at both the mRNA and the protein levels. Silencing VEGFR2 substantially enhanced phosphorylation of MEK and ERK1/2 while exerted no effects on phosphorylation of AKT, JNK and p38. Inhibiting ERK1/2 phosphorylation by LY3214996 reversed changes in VEGFR2 knockdown-induced IL-8 upregulation at the mRNA and the protein levels with no effects on cell viability. VEGFR2 overexpression significantly reduced IL-8 generation at the mRNA and the protein levels. CONCLUSION Blockade of VEGF signaling augments IL-8 secretion via MEK/ERK1/2 axis and overactivation of VEGF pathway decreases IL-8 production in hRPE cells. Upregulated IL-8 expression after VEGF signaling inhibition in hRPE cells may be responsible for being incompletely responsive to anti-VEGF remedy in neovascular AMD, and IL-8 may serve as an alternative therapeutic target for neovascular AMD.
Collapse
Affiliation(s)
- Lin-Bin Zhou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, Guangdong Province, China
| | - Ye-Qi Zhou
- Soochow University Affiliated Children's Hospital, Suzhou 215123, Jiangsu Province, China
| | - Xin-Yu Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, Guangdong Province, China
| |
Collapse
|
45
|
Terao R, Kaneko H. Lipid Signaling in Ocular Neovascularization. Int J Mol Sci 2020; 21:ijms21134758. [PMID: 32635437 PMCID: PMC7369954 DOI: 10.3390/ijms21134758] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/02/2020] [Accepted: 07/02/2020] [Indexed: 12/13/2022] Open
Abstract
Vasculogenesis and angiogenesis play a crucial role in embryonic development. Pathological neovascularization in ocular tissues can lead to vision-threatening vascular diseases, including proliferative diabetic retinopathy, retinal vein occlusion, retinopathy of prematurity, choroidal neovascularization, and corneal neovascularization. Neovascularization involves various cellular processes and signaling pathways and is regulated by angiogenic factors such as vascular endothelial growth factor (VEGF) and hypoxia-inducible factor (HIF). Modulating these circuits may represent a promising strategy to treat ocular neovascular diseases. Lipid mediators derived from membrane lipids are abundantly present in most tissues and exert a wide range of biological functions by regulating various signaling pathways. In particular, glycerophospholipids, sphingolipids, and polyunsaturated fatty acids exert potent pro-angiogenic or anti-angiogenic effects, according to the findings of numerous preclinical and clinical studies. In this review, we summarize the current knowledge regarding the regulation of ocular neovascularization by lipid mediators and their metabolites. A better understanding of the effects of lipid signaling in neovascularization may provide novel therapeutic strategies to treat ocular neovascular diseases and other human disorders.
Collapse
Affiliation(s)
- Ryo Terao
- Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
- Correspondence: ; Tel.: +81-3-3815-5411
| | - Hiroki Kaneko
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan;
| |
Collapse
|
46
|
Cytokines and Chemokines as Mediators of Prostate Cancer Metastasis. Int J Mol Sci 2020; 21:ijms21124449. [PMID: 32585812 PMCID: PMC7352203 DOI: 10.3390/ijms21124449] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 06/19/2020] [Accepted: 06/21/2020] [Indexed: 12/16/2022] Open
Abstract
The consequences of prostate cancer metastasis remain severe, with huge impact on the mortality and overall quality of life of affected patients. Despite the convoluted interplay and cross talk between various cell types and secreted factors in the metastatic process, cytokine and chemokines, along with their receptors and signaling axis, constitute important factors that help drive the sequence of events that lead to metastasis of prostate cancer. These proteins are involved in extracellular matrix remodeling, epithelial-mesenchymal-transition, angiogenesis, tumor invasion, premetastatic niche creation, extravasation, re-establishment of tumor cells in secondary organs as well as the remodeling of the metastatic tumor microenvironment. This review presents an overview of the main cytokines/chemokines, including IL-6, CXCL12, TGFβ, CXCL8, VEGF, RANKL, CCL2, CX3CL1, IL-1, IL-7, CXCL1, and CXCL16, that exert modulatory roles in prostate cancer metastasis. We also provide extensive description of their aberrant expression patterns in both advanced disease states and metastatic sites, as well as their functional involvement in the various stages of the prostate cancer metastatic process.
Collapse
|
47
|
Liang T, Xu Y, Zhu X, Zhang X, Li J, Zhao P. Aqueous humour cytokines profiles in eyes with Coats disease and the association with the severity of the disease. BMC Ophthalmol 2020; 20:178. [PMID: 32370768 PMCID: PMC7201955 DOI: 10.1186/s12886-020-01421-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 04/06/2020] [Indexed: 11/21/2022] Open
Abstract
Background To investigate aqueous humour (AH) cytokine profiles in eyes with Coats disease and analyze the association between cytokine concentrations and the severity of the disease. Methods The study included 36 patients (36 eyes) with Coats disease and 15 control patients (15 eyes) with congenital cataract. AH samples were obtained preoperatively and the concentrations of 22 different cytokines were measured through Cytometric Bead Array technology. Clinical characteristics of Coats disease, including the extent of retinal exudation and exudative retinal detachment (ERD), were recorded for analysis. Results The concentrations of 8 cytokines (VEGF, IL-6, IL-8, MCP-1, MIP-1α, IP-10, VCAM-1 and ICAM-1) were significantly higher in the Coats disease group than in the control group (all P < 0.002). Except for VCAM-1 and ICAM-1, the concentration of the other cytokines listed above showed a significant increase from stage 2 to stage 3 (all P < 0.05). Meanwhile, the concentrations of VEGF, IL-8, MCP-1 and MIP-1α showed a significant and positive association with the extent of retinal exudation and ERD (all r > 0.4, P < 0.05). Among these, IL-8 showed a strong association with the extent of retinal exudation and ERD (all r > 0.7, P < 0.001). The concentrations of IL-1α, IL-1β, IL-2, IL-4, IL-5, IL-10, IL-12, Fractalkine, RANTES, G-CSF and GM-CSF were very low in both groups. Conclusions Various cytokines in the AH, including elevated VEGF, IL-6, IL-8, MCP-1, MIP-1α, IP-10, VCAM-1 and ICAM-1, may be involved in the pathogenesis and progression of Coats disease. Increasing severity of Coats disease is significantly associated with the AH concentrations of VEGF, IL-8, MCP-1 and MIP-1α. Further clinical treatment aimed to reduce vascular leakage and antagonize neovascularization and inflammation may be useful in preventing the progression of Coats disease.
Collapse
Affiliation(s)
- Tingyi Liang
- Department of ophthalmology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yu Xu
- Department of ophthalmology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Xiuyu Zhu
- Department of ophthalmology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Xiang Zhang
- Department of ophthalmology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Jing Li
- Department of ophthalmology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Peiquan Zhao
- Department of ophthalmology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
48
|
Lam YT, Hsu CJ, Simpson PJL, Dunn LL, Chow RW, Chan KH, Yong ASC, Yu Y, Sieveking DP, Lecce L, Yuan J, Celermajer DS, Wise SG, Ng MKC. Androgens Stimulate EPC-Mediated Neovascularization and Are Associated with Increased Coronary Collateralization. Endocrinology 2020; 161:5802765. [PMID: 32157309 DOI: 10.1210/endocr/bqaa043] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 03/05/2020] [Indexed: 11/19/2022]
Abstract
Endothelial progenitor cells (EPCs) play a key role in neovascularization and have been linked to improved cardiovascular outcomes. Although there is a well-established inverse relationship between androgen levels and cardiovascular mortality in men, the role of androgens in EPC function is not fully understood. In this study, we investigated the effects of androgens on 2 subpopulations of EPCs, early EPCs (EEPCs) and late outgrowth EPCs (OECs), and their relationships with coronary collateralization. Early EPCs and OECs were isolated from the peripheral blood of young healthy men and treated with dihydrotestosterone (DHT) with or without androgen receptor (AR) antagonist, hydroxyflutamide, in vitro. Dihydrotestosterone treatment enhanced AR-mediated proliferation, migration, and tubulogenesis of EEPCs and OECs in a dose-dependent manner. Furthermore, DHT augmented EPC sensitivity to extracellular stimulation by vascular endothelial growth factor (VEGF) via increased surface VEGF receptor expression and AKT activation. In vivo, xenotransplantation of DHT pretreated human EPCs augmented blood flow recovery and angiogenesis in BALB/c nude male mice, compared to mice receiving untreated EPCs, following hindlimb ischemia. In particular, DHT pretreated human OECs exhibited higher reparative potential than EEPCs in augmenting postischemic blood flow recovery in mice. Furthermore, whole blood was collected from the coronary sinus of men with single vessel coronary artery disease (CAD) who underwent elective percutaneous intervention (n = 23). Coronary collateralization was assessed using the collateral flow index. Serum testosterone and EPC levels were measured. In men with CAD, circulating testosterone was positively associated with the extent of coronary collateralization and the levels of OECs. In conclusion, androgens enhance EPC function and promote neovascularization after ischemia in mice and are associated with coronary collateralization in men.
Collapse
Affiliation(s)
- Yuen Ting Lam
- School of Medical Science, Faculty of Health and Medicine, University of Sydney, Sydney, Australia
| | | | | | | | | | - Kim H Chan
- School of Medical Science, Faculty of Health and Medicine, University of Sydney, Sydney, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Andy S C Yong
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Young Yu
- Heart Research Institute, Sydney, Australia
| | | | | | - Jun Yuan
- Heart Research Institute, Sydney, Australia
| | - David S Celermajer
- School of Medical Science, Faculty of Health and Medicine, University of Sydney, Sydney, Australia
- Heart Research Institute, Sydney, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Steven G Wise
- School of Medical Science, Faculty of Health and Medicine, University of Sydney, Sydney, Australia
| | - Martin K C Ng
- School of Medical Science, Faculty of Health and Medicine, University of Sydney, Sydney, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia
| |
Collapse
|
49
|
Bone Marrow Stromal Cell-Derived IL-8 Upregulates PVR Expression on Multiple Myeloma Cells via NF-kB Transcription Factor. Cancers (Basel) 2020; 12:cancers12020440. [PMID: 32069911 PMCID: PMC7072437 DOI: 10.3390/cancers12020440] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/10/2020] [Accepted: 02/11/2020] [Indexed: 01/10/2023] Open
Abstract
Bone marrow stromal cells (BMSCs) strongly contribute to multiple myeloma (MM) progression, promoting the survival and growth of malignant plasma cells (PCs). However, the possible impact of these cells on the immune-mediated recognition of MM cells remains largely unknown. DNAM-1 activating receptor plays a prominent role in NK cell anti-MM response engaging the ligands poliovirus receptor (PVR) and nectin-2 on malignant PCs. Here, we analysed the role of MM patient-derived BMSCs in the regulation of PVR expression. We found that BMSCs enhance PVR surface expression on MM cells and promote their NK cell-mediated recognition. PVR upregulation occurs at transcriptional level and involves NF-kB transcription factor activation by BMSC-derived soluble factors. Indeed, overexpression of a dominant-negative mutant of IKBα blocked PVR upregulation. IL-8 plays a prominent role in these mechanisms since blockade of CXCR1/2 receptors as well as depletion of the cytokine via RNA interference prevents the enhancement of PVR expression by BMSC-derived conditioned medium. Interestingly, IL-8 is associated with stromal microvesicles which are also required for PVR upregulation via CXCR1/CXCR2 signaling activation. Our findings identify BMSCs as regulators of NK cell anti-MM response and contribute to define novel molecular pathways involved in the regulation of PVR expression in cancer cells.
Collapse
|
50
|
Zamorano P, Koning T, Oyanadel C, Mardones GA, Ehrenfeld P, Boric MP, González A, Soza A, Sánchez FA. Galectin-8 induces endothelial hyperpermeability through the eNOS pathway involving S-nitrosylation-mediated adherens junction disassembly. Carcinogenesis 2019; 40:313-323. [PMID: 30624618 DOI: 10.1093/carcin/bgz002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 12/04/2018] [Accepted: 01/04/2019] [Indexed: 12/26/2022] Open
Abstract
The permeability of endothelial cells is regulated by the stability of the adherens junctions, which is highly sensitive to kinase-mediated phosphorylation and endothelial nitric oxide synthase (eNOS)-mediated S-nitrosylation of its protein components. Solid tumors can produce a variety of factors that stimulate these signaling pathways leading to endothelial cell hyperpermeability. This generates stromal conditions that facilitate tumoral growth and dissemination. Galectin-8 (Gal-8) is overexpressed in several carcinomas and has a variety of cellular effects that can contribute to tumor pathogenicity, including angiogenesis. Here we explored whether Gal-8 has also a role in endothelial permeability. We show that recombinant Gal-8 activates eNOS, induces S-nitrosylation of p120-catenin (p120) and dissociation of adherens junction, leading to hyperpermeability of the human endothelial cell line EAhy926. This pathway involves focal-adhesion kinase (FAK) activation downstream of eNOS as a requirement for eNOS-mediated p120 S-nitrosylation. This suggests a reciprocal, yet little understood, regulation of phosphorylation and S-nitrosylation events acting upon adherens junction permeability. In addition, glutathione S-transferase (GST)-Gal-8 pull-down experiments and function-blocking β1-integrin antibodies point to β1-integrins as cell surface components involved in Gal-8-induced hyperpermeability. Endogenous Gal-8 secreted from the breast cancer cell line MCF-7 has similar hyperpermeability and signaling effects. Furthermore, the mouse cremaster model system showed that Gal-8 also activates eNOS, induces S-nitrosylation of adherens junction components and is an effective hyperpermeability agent in vivo. These results add endothelial permeability regulation by S-nitrosylation as a new function of Gal-8 that can potentially contribute to the pathogenicity of tumors overexpressing this lectin.
Collapse
Affiliation(s)
- Patricia Zamorano
- Instituto de Inmunología, Universidad Austral de Chile, Valdivia 5110566, Chile
| | - Tania Koning
- Instituto de Inmunología, Universidad Austral de Chile, Valdivia 5110566, Chile
| | - Claudia Oyanadel
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Gonzalo A Mardones
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile.,Instituto de Fisiología, Valdivia, Chile.,Centro Interdisciplinario de Estudios del Sistema Nervioso (CISNe), Valdivia, Chile
| | - Pamela Ehrenfeld
- Centro Interdisciplinario de Estudios del Sistema Nervioso (CISNe), Valdivia, Chile.,Instituto de Anatomía, Histología y Patología, Universidad Austral de Chile, Valdivia, Chile
| | | | - Alfonso González
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile.,Centro de Envejecimiento y Regeneración, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Fundación Ciencia y Vida. Santiago, Chile
| | - Andrea Soza
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile.,Centro de Envejecimiento y Regeneración, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Fabiola A Sánchez
- Instituto de Inmunología, Universidad Austral de Chile, Valdivia 5110566, Chile.,Centro Interdisciplinario de Estudios del Sistema Nervioso (CISNe), Valdivia, Chile
| |
Collapse
|