151
|
Low-Load Resistance Exercise with Blood Flow Restriction Increases Hypoxia-Induced Angiogenic Genes Expression. J Hum Kinet 2022; 84:82-91. [DOI: 10.2478/hukin-2022-0101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Abstract
The aim of the study was to determine whether low-load exercise (LL) with blood flow restriction (LL-BFR) would induce similar changes in expression of genes involved in hypoxia and angiogenesis compared to LL and high-load exercise (HL). Twenty-four males (age: 21.3 ± 1.9 years, body height: 1.74 ± 0.8 m, body mass: 73 ± 1.8 kg) were allocated into three groups: low-load exercise (LL), low-load exercise with blood-flow restriction (LL-BFR), and high-load exercise (HL). For the LL-BFR group a pneumatic cuff was inflated at 80% of the arterial occlusion pressure. All participants performed bilateral knee extension exercise, twice a week, for 8 weeks. LL and LL-BFR groups performed 3-4 sets of 15 reps at 20% 1RM, whilst the HL group performed 3-4 sets of 8-10 reps at 80% 1RM with a 60-s rest interval between sets. The hypoxia-inducible factor-1 alpha (HIF-1α) and beta (HIF-1β), vascular endothelial growth factor (VEGF), neuronal (nNOS), and inducible nitric oxide synthase (iNOS) genes expression were assessed before and after training. HIF-1α and HIF-1β mRNA levels significantly increased in the LL-BFR group and exceeded those elicited by HL and LL groups (p < .0001). VEGF gene expression was increased in both LL-BFR and HL groups, however, LL-BFR elicited a greater increase than LL (p < .0001). nNOS and iNOS genes expression significantly increased in all groups with greatest increases being observed in the LL-BFR group (p < .0001). The findings suggest that LL-BFR induces greater increases in genes expression related to hypoxia and angiogenesis than traditional resistance training.
Collapse
|
152
|
Wu J, Jin Z, Lin J, Fu Y, Wang J, Shen Y. Vessel state and immune infiltration of the angiogenesis subgroup and construction of a prediction model in osteosarcoma. Front Immunol 2022; 13:992266. [PMID: 36405691 PMCID: PMC9666676 DOI: 10.3389/fimmu.2022.992266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 10/14/2022] [Indexed: 01/25/2023] Open
Abstract
Angiogenesis has been recognized as a pivotal contributor to tumorigenesis and progression. However, the role of angiogenesis-related genes (ARGs) in vessel state, immune infiltration, and prognosis remains unknown in osteosarcoma (OS). Bulk RNA sequencing data of osteosarcoma patients were obtained from the Therapeutically Applicable Research to Generate Effective Treatments (TARGET) database, and patients were divided into two angiogenesis subgroups according to the expression of ARGs. We compared their vessel state and used two independent algorithms to evaluate the tumor microenvironment (TME) in the two subgroups. Furthermore, hub genes of differentially expressed genes (DEGs) in the two subgroups were selected to perform LASSO regression and multivariate Cox stepwise regression, and two prognostic hub genes were found. An ARG_score based on prognostic hub genes was calculated and proved to be reliable in the overall survival prediction in OS patients. Furthermore, the ARG_score was significantly associated with ARGs, immune infiltration, response to immunotherapy, and drug sensitivity. To make our prediction model perform well, clinical features were added and a highly accurate interactive nomogram was constructed. Immunohistochemistry and qRT-PCR were utilized to verify the expression of prognostic hub genes. GSE21257 from the Gene Expression Omnibus (GEO) database was used as a validation dataset to verify its robustness. In conclusion, our comprehensive analysis of angiogenesis subgroups in OS illustrated that angiogenesis may lead to different vessel states and further affect immune infiltration and prognosis of OS patients. Our findings may bring a novel perspective for the immunotherapy strategies for OS patients.
Collapse
Affiliation(s)
- Jintao Wu
- Department of Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhijian Jin
- Department of Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianwei Lin
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yucheng Fu
- Department of Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Wang
- Department of Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuhui Shen
- Department of Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
153
|
Kida Y, Yamaguchi I. The vascular protective effect of matrix Gla protein during kidney injury. FRONTIERS IN MOLECULAR MEDICINE 2022; 2:970744. [PMID: 39086959 PMCID: PMC11285670 DOI: 10.3389/fmmed.2022.970744] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 10/13/2022] [Indexed: 08/02/2024]
Abstract
Matrix Gla protein (MGP) is a small secreted protein and requires vitamin K dependent γ-carboxylation for its function. MGP has been identified as a local inhibitor of vascular calcification because MGP-deficient mice die due to severe arterial calcification and resulting arterial rupture. Clinical trials revealed that reduction in active MGP predicts poor prognosis in patients due to cardiovascular complications. However, recent studies showed that MGP controls angiogenesis during development. MGP-deficient mice demonstrated abnormal hypervascularization and arteriovenous malformations in kidneys and other organs. This abnormal angiogenesis is largely caused by excessive expression of vascular endothelial growth factor-A (VEGF-A) and VEGF receptor-2 (VEGFR2). However, only a few studies have investigated the roles of MGP in tissue injury. We observed mesangial cell proliferation and mild interstitial fibrosis in addition to increased capillaries in kidneys of MGP-null mice even without injury. We also created a mouse model with kidney injury and found that kidney damage greatly increases MGP expression in peritubular capillary endothelial cells and tubular epithelial cells. Finally, our study showed that impairment of MGP expression aggravates peritubular capillary rarefaction and accumulation of collagen-producing myofibroblasts following kidney injury. Peritubular capillary damage induces capillary loss as well as trans-differentiation of vascular pericytes into myofibroblasts. These results indicate that MGP has the vascular protective effect in the injured kidney. Clinical trials have already started to test the efficacy of MGP activation to repair vascular calcification in patients with chronic kidney diseases. In this "Hypothesis and Theory" article, we discuss possible mechanisms by which MGP protects against vascular damage during tissue injury based on our experimental results and previous results from other research groups.
Collapse
Affiliation(s)
- Yujiro Kida
- Center for Tissue and Cell Sciences, Seattle Children’s Research Institute, Seattle, WA, United States
- Department of Nephrology, Takashimadaira Chūō General Hospital, Tokyo, Japan
| | - Ikuyo Yamaguchi
- Center for Tissue and Cell Sciences, Seattle Children’s Research Institute, Seattle, WA, United States
- Division of Pediatric Nephrology and Hypertension, Department of Pediatrics, The University of Oklahoma Health Sciences Center, Oklahoma Children’s Hospital, OU Health, Oklahoma City, OK, United States
| |
Collapse
|
154
|
Rizzo-Valente VS, Fusco MA, Cruz RMML, Santos RA, Silva LS, Escaleira RC, Schulz DF, Barroso SPC, Miranda BL, Santos DZ, Gregório ML, Guerra RJA, Pavão MSG. Effects of Dermatan Sulfate from Marine Invertebrate Styela plicata in the Wound Healing Pathway: A Natural Resource Applied to Regenerative Therapy. Mar Drugs 2022; 20:676. [PMID: 36354999 PMCID: PMC9693086 DOI: 10.3390/md20110676] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/10/2022] [Accepted: 10/23/2022] [Indexed: 08/29/2023] Open
Abstract
Acute and chronic dermatological injuries need rapid tissue repair due to the susceptibility to infections. To effectively promote cutaneous wound recovery, it is essential to develop safe, low-cost, and affordable regenerative tools. Therefore, we aimed to identify the biological mechanisms involved in the wound healing properties of the glycosaminoglycan dermatan sulfate (DS), obtained from ascidian Styela plicata, a marine invertebrate, which in preliminary work from our group showed no toxicity and promoted a remarkable fibroblast proliferation and migration. In this study, 2,4-DS (50 µg/mL)-treated and control groups had the relative gene expression of 84 genes participating in the healing pathway evaluated. The results showed that 57% of the genes were overexpressed during treatment, 16% were underexpressed, and 9.52% were not detected. In silico analysis of metabolic interactions exhibited overexpression of genes related to: extracellular matrix organization, hemostasis, secretion of inflammatory mediators, and regulation of insulin-like growth factor transport and uptake. Furthermore, in C57BL/6 mice subjected to experimental wounds treated with 0.25% 2,4-DS, the histological parameters demonstrated a great capacity for vascular recovery. Additionally, this study confirmed that DS is a potent inducer of wound-healing cellular pathways and a promoter of neovascularization, being a natural ally in the tissue regeneration strategy.
Collapse
Affiliation(s)
- Vanessa S. Rizzo-Valente
- Biomedical Research Institute, Marcílio Dias Naval Hospital, Brazilian Navy, Rio de Janeiro 20725-090, Brazil
- Laboratory of Biochemistry and Cell Biology of Glycoconjugates, Clementino Fraga Filho University Hospital and Institute of Medical Biochemistry Leopoldo De Meis, Federal University of Rio de Janeiro, Rio de Janeiro 21941-913, Brazil
| | - Maria A. Fusco
- Biomedical Research Institute, Marcílio Dias Naval Hospital, Brazilian Navy, Rio de Janeiro 20725-090, Brazil
| | - Renata M. M. L. Cruz
- Biomedical Research Institute, Marcílio Dias Naval Hospital, Brazilian Navy, Rio de Janeiro 20725-090, Brazil
| | - Rachel A. Santos
- Biomedical Research Institute, Marcílio Dias Naval Hospital, Brazilian Navy, Rio de Janeiro 20725-090, Brazil
| | - Lucas S. Silva
- Biomedical Research Institute, Marcílio Dias Naval Hospital, Brazilian Navy, Rio de Janeiro 20725-090, Brazil
| | - Roberta C. Escaleira
- Biomedical Research Institute, Marcílio Dias Naval Hospital, Brazilian Navy, Rio de Janeiro 20725-090, Brazil
| | - Daniel F. Schulz
- Biomedical Research Institute, Marcílio Dias Naval Hospital, Brazilian Navy, Rio de Janeiro 20725-090, Brazil
| | - Shana P. C. Barroso
- Biomedical Research Institute, Marcílio Dias Naval Hospital, Brazilian Navy, Rio de Janeiro 20725-090, Brazil
| | - Bruno L. Miranda
- Biomedical Research Institute, Marcílio Dias Naval Hospital, Brazilian Navy, Rio de Janeiro 20725-090, Brazil
| | - Daniela Z. Santos
- Biomedical Research Institute, Marcílio Dias Naval Hospital, Brazilian Navy, Rio de Janeiro 20725-090, Brazil
| | - Marcelo L. Gregório
- Biomedical Research Institute, Marcílio Dias Naval Hospital, Brazilian Navy, Rio de Janeiro 20725-090, Brazil
| | - Rodrigo J. A. Guerra
- Biomedical Research Institute, Marcílio Dias Naval Hospital, Brazilian Navy, Rio de Janeiro 20725-090, Brazil
| | - Mauro S. G. Pavão
- Laboratory of Biochemistry and Cell Biology of Glycoconjugates, Clementino Fraga Filho University Hospital and Institute of Medical Biochemistry Leopoldo De Meis, Federal University of Rio de Janeiro, Rio de Janeiro 21941-913, Brazil
| |
Collapse
|
155
|
Single-cell sequencing deconvolutes cellular responses to exercise in human skeletal muscle. Commun Biol 2022; 5:1121. [PMID: 36273106 PMCID: PMC9588010 DOI: 10.1038/s42003-022-04088-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 10/11/2022] [Indexed: 11/08/2022] Open
Abstract
Skeletal muscle adaptations to exercise have been associated with a range of health-related benefits, but cell type-specific adaptations within the muscle are incompletely understood. Here we use single-cell sequencing to determine the effects of exercise on cellular composition and cell type-specific processes in human skeletal muscle before and after intense exercise. Fifteen clusters originating from six different cell populations were identified. Most cell populations remained quantitatively stable after exercise, but a large transcriptional response was observed in mesenchymal, endothelial, and myogenic cells, suggesting that these cells are specifically involved in skeletal muscle remodeling. We found three subpopulations of myogenic cells characterized by different maturation stages based on the expression of markers such as PAX7, MYOD1, TNNI1, and TNNI2. Exercise accelerated the trajectory of myogenic progenitor cells towards maturation by increasing the transcriptional features of fast- and slow-twitch muscle fibers. The transcriptional regulation of these contractile elements upon differentiation was validated in vitro on primary myoblast cells. The cell type-specific adaptive mechanisms induced by exercise presented here contribute to the understanding of the skeletal muscle adaptations triggered by physical activity and may ultimately have implications for physiological and pathological processes affecting skeletal muscle, such as sarcopenia, cachexia, and glucose homeostasis. Single-cell RNA-sequencing of human skeletal muscle before and after exercise highlights how physical activity changes the composition and transcriptomic profile of muscle tissue.
Collapse
|
156
|
Zhang Z, Warner KA, Mantesso A, Nör JE. PDGF-BB signaling via PDGFR-β regulates the maturation of blood vessels generated upon vasculogenic differentiation of dental pulp stem cells. Front Cell Dev Biol 2022; 10:977725. [PMID: 36340037 PMCID: PMC9627550 DOI: 10.3389/fcell.2022.977725] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 09/29/2022] [Indexed: 11/13/2022] Open
Abstract
A functional vascular network requires that blood vessels are invested by mural cells. We have shown that dental pulp stem cells (DPSC) can undergo vasculogenic differentiation, and that the resulting vessels anastomize with the host vasculature and become functional (blood carrying) vessels. However, the mechanisms underlying the maturation of DPSC-derived blood vessels remains unclear. Here, we performed a series of studies to understand the process of mural cell investment of blood vessels generated upon vasculogenic differentiation of dental pulp stem cells. Primary human DPSC were co-cultured with primary human umbilical artery smooth muscle cells (HUASMC) in 3D gels in presence of vasculogenic differentiation medium. We observed DPSC capillary sprout formation and SMC recruitment, alignment and remodeling that resulted in complex vascular networks. While HUASMC enhanced the number of capillary sprouts and stabilized the capillary network when co-cultured with DPSC, HUASMC by themselves were unable to form capillary sprouts. In vivo, GFP transduced human DPSC seeded in biodegradable scaffolds and transplanted into immunodeficient mice generated functional human blood vessels invested with murine smooth muscle actin (SMA)-positive, GFP-negative cells. Inhibition of PDGFR-β signaling prevented the SMC investment of DPSC-derived capillary sprouts in vitro and of DPSC-derived blood vessels in vivo. In contrast, inhibition of Tie-2 signaling did not have a significant effect on the SMC recruitment in DPSC-derived vascular structures. Collectively, these results demonstrate that PDGF-BB signaling via PDGFR-β regulates the process of maturation (mural investment) of blood vessels generated upon vasculogenic differentiation of human dental pulp stem cells.
Collapse
Affiliation(s)
- Zhaocheng Zhang
- Angiogenesis Research Laboratory, Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI, United States
| | - Kristy A. Warner
- Angiogenesis Research Laboratory, Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI, United States
| | - Andrea Mantesso
- Angiogenesis Research Laboratory, Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI, United States
| | - Jacques E. Nör
- Angiogenesis Research Laboratory, Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI, United States
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI, United States
- Department of Otolaryngology, University of Michigan School of Medicine, Ann Arbor, MI, United States
| |
Collapse
|
157
|
Maiorano BA, Parisi A, Maiello E, Ciardiello D. The Interplay between Anti-Angiogenics and Immunotherapy in Colorectal Cancer. LIFE (BASEL, SWITZERLAND) 2022; 12:life12101552. [PMID: 36294987 PMCID: PMC9604892 DOI: 10.3390/life12101552] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 09/30/2022] [Accepted: 10/02/2022] [Indexed: 11/06/2022]
Abstract
Simple Summary Colorectal cancer is a frequent and lethal neoplasm. The tumor often creates new vessels to grow and spread—a process called ‘angiogenesis’. Therefore, drugs blocking angiogenesis are effective against this malignancy. On the other side, immune checkpoint inhibitors, which unleash the immune system to fight against tumors, have limited efficacy in patients carrying instability of DNA regions called microsatellites. However, there is an interaction between angiogenic factors and the immune system. This gives a chance to combine anti-angiogenic agents and immune checkpoint inhibitors to improve the efficacy of treating this malignancy. Abstract Angiogenesis, a hallmark of cancer, plays a fundamental role in colorectal cancer (CRC). Anti-angiogenic drugs and chemotherapy represent a standard of care for treating metastatic disease. Immune checkpoint inhibitors (ICIs) have changed the therapeutic algorithm of many solid tumors. However, the efficacy of ICIs is limited to mCRC patients carrying microsatellite instability (MSI-H), which represent approximately 3–5% of mCRC. Emerging evidence suggests that anti-angiogenic drugs could exhibit immunomodulatory properties. Thus, there is a strong rationale for combining anti-angiogenics and ICIs to improve efficacy in the metastatic setting. Our review summarizes the pre-clinical and clinical evidence regarding the combination of anti-angiogenics and ICIs in mCRC to deepen the possible application in daily clinical practice.
Collapse
Affiliation(s)
- Brigida Anna Maiorano
- Oncology Unit, IRCCS Foundation Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
- Correspondence:
| | - Alessandro Parisi
- Department of Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria, Ospedali Riuniti di Ancona, 60126 Ancona, Italy
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Evaristo Maiello
- Oncology Unit, IRCCS Foundation Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Davide Ciardiello
- Oncology Unit, IRCCS Foundation Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
- Medical Oncology Unit, Department of Precision Medicine, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy
| |
Collapse
|
158
|
Barć P, Antkiewicz M, Frączkowska-Sioma K, Kupczyńska D, Lubieniecki P, Witkiewicz W, Małodobra-Mazur M, Baczyńska D, Janczak D, Skóra JP. Two-Stage Gene Therapy (VEGF, HGF and ANG1 Plasmids) as Adjunctive Therapy in the Treatment of Critical Lower Limb Ischemia in Diabetic Foot Syndrome. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:12818. [PMID: 36232122 PMCID: PMC9564889 DOI: 10.3390/ijerph191912818] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/03/2022] [Accepted: 10/04/2022] [Indexed: 06/16/2023]
Abstract
One of the most serious problems in people with diabetes is diabetic foot syndrome. Due to the peripheral location of atherosclerotic lesions in the arterial system of the lower extremities, endovascular treatment plays a dominant role. However, carrying out these procedures is not always possible and does not always bring the expected results. Gene therapy, which stimulates angiogenesis, improves not only the inflow from the proximal limb but also the blood redistribution in individual angiosomes. Due to the encouraging results of sequential treatment consisting of intramuscular injections of VEGF/HGF bicistronic plasmids followed by a month of ANG1 plasmids, we decided to use the described method for the treatment of critical ischemia of the lower limbs in the course of diabetes and, more specifically, in diabetic foot syndrome. Twenty-four patients meeting the inclusion criteria were enrolled in the study. They were randomly divided into two equal groups. The first group of patients was subjected to gene therapy, where the patients received intramuscular injections of pIRES/VEGF165/HGF plasmids and 1 month of ANG-1 plasmids. The remaining patients constituted the control group. Gene therapy was well tolerated by most patients. The wounds healed significantly better in Group 1. The minimal value of ABI increased significantly in Group 1 from 0.44 ± 0.14 (± standard deviation) to 0.47 ± 0.12 (with p = 0.028) at the end of the study. There were no significant differences in the control group. In the gene treatment group, PtcO2 increased significantly (from 28.71 ± 10.89 mmHg to 33.9 ± 6.33 mmHg with p = 0.001), while in Group 2, no statistically significant changes were found. The observed resting pain decreased significantly in both groups (Group 1 decreased from 6.80 ± 1.48 to 2.10 ± 1.10; p < 0.001; the control group decreased from 7.44 ± 1.42 to 3.78 ± 1.64 with p < 0.001). In our study, we evaluated the effectiveness of gene therapy with the growth factors described above in patients with CLI in the course of complicated DM. The therapy was shown to be effective with minimal side effects. No serious complications were observed.
Collapse
Affiliation(s)
- Piotr Barć
- Department and Clinic of Vascular, General and Transplantation Surgery, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland
| | - Maciej Antkiewicz
- Department and Clinic of Vascular, General and Transplantation Surgery, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland
| | - Katarzyna Frączkowska-Sioma
- Department and Clinic of Vascular, General and Transplantation Surgery, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland
| | - Diana Kupczyńska
- Department and Clinic of Vascular, General and Transplantation Surgery, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland
| | - Paweł Lubieniecki
- Department of Angiology, Hypertension and Diabetology, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland
| | - Wojciech Witkiewicz
- Research and Development Center, Regional Specialized Hospital in Wroclaw, Kamienskiego 73a, 51-124 Wroclaw, Poland
| | - Małgorzata Małodobra-Mazur
- Department of Forensic Medicine, Division of Molecular Techniques, Wroclaw Medical University, M. Curie-Sklodowskiej 52, 50-369 Wroclaw, Poland
| | - Dagmara Baczyńska
- Department of Molecular and Cellular Biology, Wroclaw Medical University, Borowska 211A, 50-556 Wrocław, Poland
| | - Dariusz Janczak
- Department and Clinic of Vascular, General and Transplantation Surgery, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland
| | - Jan Paweł Skóra
- Department and Clinic of Vascular, General and Transplantation Surgery, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland
| |
Collapse
|
159
|
Husain A, Bhutani M, Parveen S, Khan SA, Ahmad A, Iqbal MA. Design, Synthesis, In Vitro Cytotoxicity, ADME Prediction, and Molecular Docking Study of Benzimidazole-Linked Pyrrolone and N-Benzylpyrrolone Derivatives. RUSSIAN JOURNAL OF ORGANIC CHEMISTRY 2022. [DOI: 10.1134/s1070428022100098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
160
|
Dimerization of the C-type lectin-like receptor CD93 promotes its binding to Multimerin-2 in endothelial cells. Int J Biol Macromol 2022; 224:453-464. [DOI: 10.1016/j.ijbiomac.2022.10.136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 09/30/2022] [Accepted: 10/15/2022] [Indexed: 11/05/2022]
|
161
|
Kokai D, Stanic B, Tesic B, Samardzija Nenadov D, Pogrmic-Majkic K, Fa Nedeljkovic S, Andric N. Dibutyl phthalate promotes angiogenesis in EA.hy926 cells through estrogen receptor-dependent activation of ERK1/2, PI3K-Akt, and NO signaling pathways. Chem Biol Interact 2022; 366:110174. [PMID: 36089060 DOI: 10.1016/j.cbi.2022.110174] [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: 04/11/2022] [Revised: 08/11/2022] [Accepted: 09/05/2022] [Indexed: 11/03/2022]
Abstract
Dibutyl phthalate (DBP) is an endocrine disruptor that has been widely used in various products of human use. DBP exposure has been associated with reproductive and cardiovascular diseases and metabolic disorders. Although dysfunction of the vascular endothelium is responsible for many cardiovascular and metabolic diseases, little is known about the effects of DBP on human endothelium. In this study, we investigated the effect of three concentrations of DBP (10-6, 10-5, and 10-4 M) on angiogenesis in human endothelial cell (EC) line EA.hy926 after acute exposure. Tube formation assay was used to investigate in vitro angiogenesis, whereas qRT-PCR was employed to measure mRNA expression. The effect of DBP on extracellular signal-regulated kinase 1/2 (ERK1/2), phosphatidylinositol 3-kinase (PI3K)-protein kinase B (Akt), and endothelial nitric oxide (NO) synthase (eNOS) activation was examined using Western blotting, whereas the Griess method was used to assess NO production. Results show that the 24-h-long exposure to 10-4 M DBP increased endothelial tube formation, which was prevented by addition of U0126 (ERK1/2 inhibitor), wortmannin (PI3K-Akt inhibitor), and l-NAME (NOS inhibitor). Short exposure to 10-4 M DBP (from 15 to 120 min) phosphorylated ERK1/2, Akt, and eNOS in different time points and increased NO production after 24 and 48 h of exposure. Application of nuclear estrogen receptor (ER) and G protein-coupled ER (GPER) inhibitors ICI 182,780 and G-15, respectively, abolished the DBP-mediated ERK1/2, Akt, and eNOS phosphorylation and increase in NO production. In this study, we report for the first time that DBP exerts a pro-angiogenic effect on human vascular ECs and describe the molecular mechanism involving ER- and GPER-dependent activation of ERK1/2, PI3K-Akt, and NO signaling pathways.
Collapse
Affiliation(s)
- Dunja Kokai
- University of Novi Sad, Faculty of Sciences, Department of Biology and Ecology, Serbia
| | - Bojana Stanic
- University of Novi Sad, Faculty of Sciences, Department of Biology and Ecology, Serbia.
| | - Biljana Tesic
- University of Novi Sad, Faculty of Sciences, Department of Biology and Ecology, Serbia
| | | | | | | | - Nebojsa Andric
- University of Novi Sad, Faculty of Sciences, Department of Biology and Ecology, Serbia
| |
Collapse
|
162
|
Zhou T, Li Y, Zhang H, Pan L, Pang J, Yuan Q, Li G, Jie L, Wang Y, Zhang Y. 4-(2-Butyl-6,7-dichloro-2-cyclopentyl-indan-1-on-5-yl) oxobutyric acid inhibits angiogenesis via modulation of vascular endothelial growth factor receptor 2 signaling pathway. Front Cardiovasc Med 2022; 9:969616. [PMID: 36211567 PMCID: PMC9537693 DOI: 10.3389/fcvm.2022.969616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/15/2022] [Indexed: 11/17/2022] Open
Abstract
4-(2-Butyl-6,7-dichloro-2-cyclopentyl-indan-1-on-5-yl) oxobutyric acid (DCPIB), was discovered to be a potent and specific antagonist of volume-regulated anion channel that is closely linked to angiogenesis. However, the effect of DCPIB on angiogenesis remains unclear. Here, we found that DCPIB inhibited angiogenesis in the corneal suture and myocardial infarction in vivo model. In addition, DCPIB inhibited human umbilical vein endothelial cell migration, tube formation and proliferation in vitro. Moreover, DCPIB repressed the activation and expression of vascular endothelial growth factor receptor 2 (VEGFR2) and its downstream signaling pathway. Computer modeling further confirmed that DCPIB binds with high affinity to VEGFR2. Collectively, we present evidence supporting an antiangiogenic role of DCPIB by targeting VEGFR2 signaling pathway, which suggests that DCPIB is a valuable lead compound for the treatment of angiogenesis-related diseases.
Collapse
Affiliation(s)
- Tianli Zhou
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yunda Li
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Heqiang Zhang
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Lei Pan
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jinglong Pang
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Qian Yuan
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Guiyang Li
- Department of Cardiology, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Lingjun Jie
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Department of Cardiology, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- *Correspondence: Lingjun Jie
| | - Yan Wang
- Department of Cardiology, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Yan Wang
| | - Yanhui Zhang
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Yanhui Zhang
| |
Collapse
|
163
|
Metabolic Reprogramming in Tumor Endothelial Cells. Int J Mol Sci 2022; 23:ijms231911052. [PMID: 36232355 PMCID: PMC9570383 DOI: 10.3390/ijms231911052] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/15/2022] [Accepted: 09/17/2022] [Indexed: 11/29/2022] Open
Abstract
The dynamic crosstalk between the different components of the tumor microenvironment is critical to determine cancer progression, metastatic dissemination, tumor immunity, and therapeutic responses. Angiogenesis is critical for tumor growth, and abnormal blood vessels contribute to hypoxia and acidosis in the tumor microenvironment. In this hostile environment, cancer and stromal cells have the ability to alter their metabolism in order to support the high energetic demands and favor rapid tumor proliferation. Recent advances have shown that tumor endothelial cell metabolism is reprogrammed, and that targeting endothelial metabolic pathways impacts developmental and pathological vessel sprouting. Therefore, the use of metabolic antiangiogenic therapies to normalize the blood vasculature, in combination with immunotherapies, offers a clinical niche to treat cancer.
Collapse
|
164
|
Subramanian A, Zakeri P, Mousa M, Alnaqbi H, Alshamsi FY, Bettoni L, Damiani E, Alsafar H, Saeys Y, Carmeliet P. Angiogenesis goes computational - The future way forward to discover new angiogenic targets? Comput Struct Biotechnol J 2022; 20:5235-5255. [PMID: 36187917 PMCID: PMC9508490 DOI: 10.1016/j.csbj.2022.09.019] [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: 07/08/2022] [Revised: 09/09/2022] [Accepted: 09/09/2022] [Indexed: 11/26/2022] Open
Abstract
Multi-omics technologies are being increasingly utilized in angiogenesis research. Yet, computational methods have not been widely used for angiogenic target discovery and prioritization in this field, partly because (wet-lab) vascular biologists are insufficiently familiar with computational biology tools and the opportunities they may offer. With this review, written for vascular biologists who lack expertise in computational methods, we aspire to break boundaries between both fields and to illustrate the potential of these tools for future angiogenic target discovery. We provide a comprehensive survey of currently available computational approaches that may be useful in prioritizing candidate genes, predicting associated mechanisms, and identifying their specificity to endothelial cell subtypes. We specifically highlight tools that use flexible, machine learning frameworks for large-scale data integration and gene prioritization. For each purpose-oriented category of tools, we describe underlying conceptual principles, highlight interesting applications and discuss limitations. Finally, we will discuss challenges and recommend some guidelines which can help to optimize the process of accurate target discovery.
Collapse
Affiliation(s)
- Abhishek Subramanian
- Laboratory of Angiogenesis & Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Angiogenesis & Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Pooya Zakeri
- Laboratory of Angiogenesis & Vascular Heterogeneity, Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Centre for Brain and Disease Research, Flanders Institute for Biotechnology (VIB), Leuven, Belgium
- Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Mira Mousa
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Halima Alnaqbi
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Fatima Yousif Alshamsi
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Leo Bettoni
- Laboratory of Angiogenesis & Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Angiogenesis & Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Ernesto Damiani
- Robotics and Intelligent Systems Institute, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Habiba Alsafar
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Yvan Saeys
- Data Mining and Modelling for Biomedicine Group, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis & Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Angiogenesis & Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium
- Laboratory of Angiogenesis & Vascular Heterogeneity, Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| |
Collapse
|
165
|
Li G, Wang Q, Feng J, Wang J, Wang Y, Huang X, Shao T, Deng X, Cao Y, Zhou M, Zhao C. Recent insights into the role of defensins in diabetic wound healing. Biomed Pharmacother 2022; 155:113694. [PMID: 36099789 DOI: 10.1016/j.biopha.2022.113694] [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: 07/13/2022] [Revised: 09/07/2022] [Accepted: 09/09/2022] [Indexed: 11/28/2022] Open
Abstract
Diabetic wound, one of the most common serious complications of diabetic patients, is an important factor in disability and death. Much of the research on the pathophysiology of diabetic wound healing has long focused on mechanisms mediated by hyperglycemia, chronic inflammation, microcirculatory and macrocirculatory dysfunction. However, recent evidence suggests that defensins may play a crucial role in the development and perpetuation of diabetic wound healing. The available findings suggest that defensins exert a beneficial influence on diabetic wound healing through antimicrobial, immunomodulatory, angiogenic, tissue regenerator effects, and insulin resistance improvement. Therefore, summarizing the existing research progress on defensins in the diabetic wound may present a promising strategy for diabetic patients.
Collapse
Affiliation(s)
- Gen Li
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China; Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qixue Wang
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China; Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiawei Feng
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China; Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jialin Wang
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China; Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yuqing Wang
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China; Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiaoting Huang
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China; Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Tengteng Shao
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Xiaofei Deng
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Yemin Cao
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Mingmei Zhou
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China; Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Cheng Zhao
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China.
| |
Collapse
|
166
|
Zhang J, Wang X, Lin Z, Zhu T. Pro-angiogenic activity of isofuran. Biosci Biotechnol Biochem 2022; 86:1506-1514. [PMID: 36066914 DOI: 10.1093/bbb/zbac146] [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: 06/05/2022] [Accepted: 08/29/2022] [Indexed: 11/12/2022]
Abstract
Isofurans (IsoFs) are a series of novel discovered lipid peroxidation products. This study focused on the investigation of angiogenic property of IsoF. MTT stain assay indicated that 1 μM IsoF had the most bioactivity in RBECs. IsoF significantly promoted cellular proliferation and migration and remarkably decreased staurosporine-induced apoptosis by TUNEL assay in the RBECs. It successfully up-regulated rat aortic vascularization and choroid explant sprouting, extracellular regulated protein kinases (ERK)1/2, and triggered calcium release. RT-PCR examination indicated that IsoF up-regulated tumor necrosis factor (TNF)α, angiopoietin-1 receptor (Tie2), and vascular endothelial growth factor (VEGF)-A, but did not interfere with caspase 2 and VEGF-C in the RBECs. IsoF has pro-angiogenic activity. Calcium release and ERK1/2 phosphorylation may be involved in the signaling of the IsoF-induced up-regulation of TNFα, Tie2, and VEGF-A, which could be the molecular mechanism of the pro-angiogenic activity of the IsoF.
Collapse
Affiliation(s)
- Jingxia Zhang
- Key Laboratory of Translational Tumor Medicine in Fujian Province, School of Basic Medical Science Putian University, Putian, Fujian, China
| | - Xiangjiang Wang
- Key Laboratory of Translational Tumor Medicine in Fujian Province, School of Basic Medical Science Putian University, Putian, Fujian, China
| | - Zhiping Lin
- Key Laboratory of Translational Tumor Medicine in Fujian Province, School of Basic Medical Science Putian University, Putian, Fujian, China
| | - Tang Zhu
- Key Laboratory of Translational Tumor Medicine in Fujian Province, School of Basic Medical Science Putian University, Putian, Fujian, China.,Yujia Biotech., Guangzhou, Guangdong, China
| |
Collapse
|
167
|
Saeed MU, Hussain N, Shahbaz A, Hameed T, Iqbal HMN, Bilal M. Bioprospecting microalgae and cyanobacteria for biopharmaceutical applications. J Basic Microbiol 2022; 62:1110-1124. [PMID: 34914840 DOI: 10.1002/jobm.202100445] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 11/19/2021] [Accepted: 11/27/2021] [Indexed: 02/05/2023]
Abstract
Microalgae and cyanobacteria have sparked a lot of interest due to their potential in various industries like biorefineries, biopharmaceuticals, food supplements, nutraceuticals, and other high-value products. Polysaccharides, vitamins, proteins, enzymes, and steroids are valuable products isolated from microalgae and cyanobacteria and potentially used in health and biomedical applications. Bioactive compounds derived from microalgae and cyanobacteria exhibit various pharmaceutical properties like antibacterial, anticancer, antiviral, antialgal, and antioxidant. From the properties listed above, the research for novel antibiotics has become particularly appropriate. In addition, the possible emergence of resistance against pathogens, as well as the potential decline in antibiotic efficacy, has prompted researchers to look for a new source of antibiotics. Microalgae and cyanobacteria have indicated a great and unexplored potential among these sources. For this reason, microalgae and cyanobacteria have been highlighted for their efficiency in different industrial sectors, as well as for their potential uses in the betterment of human and environmental health. This review gives an overview of bioactive compounds and metabolites with several biological properties isolated from microalgae and cyanobacteria for treating different animal and human diseases.
Collapse
Affiliation(s)
- Muhammad U Saeed
- Center for Applied Molecular Biology (CAMB), University of the Punjab, Lahore, Pakistan
| | - Nazim Hussain
- Center for Applied Molecular Biology (CAMB), University of the Punjab, Lahore, Pakistan
| | - Areej Shahbaz
- Center for Applied Molecular Biology (CAMB), University of the Punjab, Lahore, Pakistan
| | - Tooba Hameed
- School of Biochemistry & Biotechnology, University of the Punjab Lahore, Lahore, Pakistan
| | - Hafiz M N Iqbal
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey, Mexico
| | - Muhammad Bilal
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian, China
| |
Collapse
|
168
|
Hashemi G, Dight J, Khosrotehrani K, Sormani L. Melanoma Tumour Vascularization and Tissue-Resident Endothelial Progenitor Cells. Cancers (Basel) 2022; 14:4216. [PMID: 36077754 PMCID: PMC9454996 DOI: 10.3390/cancers14174216] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/17/2022] [Accepted: 08/24/2022] [Indexed: 11/20/2022] Open
Abstract
The aggressiveness of solid cancers, such as melanoma, relies on their metastatic potential. It has become evident that this key cause of mortality is largely conferred by the tumour-associated stromal cells, especially endothelial cells. In addition to their essential role in the formation of the tumour vasculature, endothelial cells significantly contribute to the establishment of the tumour microenvironment, thus enabling the dissemination of cancer cells. Melanoma tumour vascularization occurs through diverse biological processes. Vasculogenesis is the formation of de novo blood vessels from endothelial progenitor cells (EPCs), and recent research has shown the role of EPCs in melanoma tumour vascularization. A more detailed understanding of the complex role of EPCs and how they contribute to the abnormal vessel structures in tumours is of importance. Moreover, anti-angiogenic drugs have a limited effect on melanoma tumour vascularization, and the role of these drugs on EPCs remains to be clarified. Overall, targeting cancer vasculature remains a challenge, and the role of anti-angiogenic drugs and combination therapies in melanoma, a focus of this review, is an area of extensive exploration.
Collapse
Affiliation(s)
| | | | - Kiarash Khosrotehrani
- Experimental Dermatology Group, Dermatology Research Centre, The UQ Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Laura Sormani
- Experimental Dermatology Group, Dermatology Research Centre, The UQ Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| |
Collapse
|
169
|
Koning M, Dumas SJ, Avramut MC, Koning RI, Meta E, Lievers E, Wiersma LE, Borri M, Liang X, Xie L, Liu P, Chen F, Lin L, Luo Y, Mulder J, Spijker HS, Jaffredo T, van den Berg BM, Carmeliet P, van den Berg CW, Rabelink TJ. Vasculogenesis in kidney organoids upon transplantation. NPJ Regen Med 2022; 7:40. [DOI: 10.1038/s41536-022-00237-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 07/20/2022] [Indexed: 02/07/2023] Open
Abstract
AbstractHuman induced pluripotent stem cell-derived kidney organoids have potential for disease modeling and to be developed into clinically transplantable auxiliary tissue. However, they lack a functional vasculature, and the sparse endogenous endothelial cells (ECs) are lost upon prolonged culture in vitro, limiting maturation and applicability. Here, we use intracoelomic transplantation in chicken embryos followed by single-cell RNA sequencing and advanced imaging platforms to induce and study vasculogenesis in kidney organoids. We show expansion of human organoid-derived ECs that reorganize into perfused capillaries and form a chimeric vascular network with host-derived blood vessels. Ligand-receptor analysis infers extensive potential interactions of human ECs with perivascular cells upon transplantation, enabling vessel wall stabilization. Perfused glomeruli display maturation and morphogenesis to capillary loop stage. Our findings demonstrate the beneficial effect of vascularization on not only epithelial cell types, but also the mesenchymal compartment, inducing the expansion of ´on target´ perivascular stromal cells, which in turn are required for further maturation and stabilization of the neo-vasculature. The here described vasculogenic capacity of kidney organoids will have to be deployed to achieve meaningful glomerular maturation and kidney morphogenesis in vitro.
Collapse
|
170
|
Sema4C Is Required for Vascular and Primary Motor Neuronal Patterning in Zebrafish. Cells 2022; 11:cells11162527. [PMID: 36010604 PMCID: PMC9406964 DOI: 10.3390/cells11162527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/29/2022] [Accepted: 08/04/2022] [Indexed: 12/03/2022] Open
Abstract
Endothelial cells (ECs) and neurons share a number of common signaling pathways and molecular mediators to orchestrate directional migration and guide the pattern of the vascular network and nervous system. So far, research concerning the functional coupling between vascular and neuronal pathfinding remains insufficient. Semaphorin4C (sema4C), a member of class 4 semaphorins, is initially described in the nervous system, whose role has been demonstrated in diverse biological developments. The present study focused on the role of sema4C in the vascular and neural development process in zebrafish embryos. It confirmed that sema4C is expressed in both the nervous system and intersegmental vessels (ISVs) in zebrafish embryos by diverse expression analysis. It also showed that the knockdown of sema4C caused a serious pathfinding anomaly both in the ISVs and primary motor neurons (PMNs) of zebrafish embryos. In addition, overexpressing exogenous sema4C mRNA in sema4C morphants remarkably neutralized the defective pattern of the vascular and neural system. Collectively, this report suggests that sema4C acts as a dual guiding factor regulating vascular and neuronal development. These findings elucidate a new molecular mechanism underlying blood vessel and nerve development and might serve as groundwork for future research on functional coupling between both systems.
Collapse
|
171
|
Lee HS, Song HJ, Park Y, Smolensky D, Lee SH. Permethrin inhibits tube formation and viability of endothelial cells. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2022; 102:4079-4085. [PMID: 34997580 DOI: 10.1002/jsfa.11757] [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: 08/26/2021] [Revised: 11/11/2021] [Accepted: 12/22/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND Exposure to environmental chemicals has been linked with endothelial dysfunction, which is a leading cause of human diseases, including atherosclerosis. Permethrin is a frequently used synthetic pyrethroid insecticide for which longer exposure may cause toxicity in several types of tissues and the development of metabolic diseases, including atherosclerosis, obesity and diabetes. The present study was designed to evaluate the potential adverse effect of permethrin on the function and activity of human endothelial cells. RESULTS Permethrin was found to repress migration and tube formation by human umbilical vein endothelial cells (HUVECs) in a dose-dependent manner, as well as to significantly repress their viability after 24 and 48 h of treatment. Furthermore, increased reactive oxygen species (ROS) production was observed in cells treated with permethrin, and the permethrin-induced repression of cell viability was ROS-dependent. Permethrin did not influence apoptosis, necrosis or mitochondrial membrane potential in HUVECs. CONCLUSION The results of the present study suggest that permethrin represses angiogenesis and viability through ROS-dependent and cell growth-, apoptosis- and necrosis-independent means. © 2022 Society of Chemical Industry.
Collapse
Affiliation(s)
- Hee-Seop Lee
- Department of Nutrition and Food Science, College of Agriculture and Natural Resources, University of Maryland, College Park, MD, USA
| | - Hee-Jung Song
- Department of Nutrition and Food Science, College of Agriculture and Natural Resources, University of Maryland, College Park, MD, USA
| | - Yeonhwa Park
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
| | - Dmitriy Smolensky
- Grain Quality and Structure Research Unit, Agricultural Research Service, U.S. Department of Agriculture, Manhattan, KS, USA
| | - Seong-Ho Lee
- Department of Nutrition and Food Science, College of Agriculture and Natural Resources, University of Maryland, College Park, MD, USA
| |
Collapse
|
172
|
Impact of Non-Pharmacological Interventions on the Mechanisms of Atherosclerosis. Int J Mol Sci 2022; 23:ijms23169097. [PMID: 36012362 PMCID: PMC9409393 DOI: 10.3390/ijms23169097] [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: 07/29/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 11/30/2022] Open
Abstract
Atherosclerosis remains the leading cause of mortality and morbidity worldwide characterized by the deposition of lipids and fibrous elements in the form of atheroma plaques in vascular areas which are hemodynamically overloaded. The global burden of atherosclerotic cardiovascular disease is steadily increasing and is considered the largest known non-infectious pandemic. The management of atherosclerotic cardiovascular disease is increasing the cost of health care worldwide, which is a concern for researchers and physicians and has caused them to strive to find effective long-term strategies to improve the efficiency of treatments by managing conventional risk factors. Primary prevention of atherosclerotic cardiovascular disease is the preferred method to reduce cardiovascular risk. Fasting, a Mediterranean diet, and caloric restriction can be considered useful clinical tools. The protective impact of physical exercise over the cardiovascular system has been studied in recent years with the intention of explaining the mechanisms involved; the increase in heat shock proteins, antioxidant enzymes and regulators of cardiac myocyte proliferation concentration seem to be the molecular and biochemical shifts that are involved. Developing new therapeutic strategies such as vagus nerve stimulation, either to prevent or slow the disease’s onset and progression, will surely have a profound effect on the lives of millions of people.
Collapse
|
173
|
Mumu M, Das A, Emran TB, Mitra S, Islam F, Roy A, Karim MM, Das R, Park MN, Chandran D, Sharma R, Khandaker MU, Idris AM, Kim B. Fucoxanthin: A Promising Phytochemical on Diverse Pharmacological Targets. Front Pharmacol 2022; 13:929442. [PMID: 35983376 PMCID: PMC9379326 DOI: 10.3389/fphar.2022.929442] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 05/23/2022] [Indexed: 12/24/2022] Open
Abstract
Fucoxanthin (FX) is a special carotenoid having an allenic bond in its structure. FX is extracted from a variety of algae and edible seaweeds. It has been proved to contain numerous health benefits and preventive effects against diseases like diabetes, obesity, liver cirrhosis, malignant cancer, etc. Thus, FX can be used as a potent source of both pharmacological and nutritional ingredient to prevent infectious diseases. In this review, we gathered the information regarding the current findings on antimicrobial, antioxidant, anti-inflammatory, skin protective, anti-obesity, antidiabetic, hepatoprotective, and other properties of FX including its bioavailability and stability characteristics. This review aims to assist further biochemical studies in order to develop further pharmaceutical assets and nutritional products in combination with FX and its various metabolites.
Collapse
Affiliation(s)
- Mumtaza Mumu
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Ayan Das
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, Bangladesh
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
- *Correspondence: Talha Bin Emran, ; Abubakr M. Idris, ; Bonglee Kim,
| | - Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, Bangladesh
| | - Fahadul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Arpita Roy
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, India
| | - Md. Mobarak Karim
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | - Rajib Das
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Moon Nyeo Park
- Department of Pathology College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Deepak Chandran
- Department of Veterinary Sciences and Animal Husbandry, Amrita School of Agricultural Sciences, Amrita Vishwa Vidyapeetham University, Coimbatore, India
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Mayeen Uddin Khandaker
- Centre for Applied Physics and Radiation Technologies, School of Engineering and Technology, Sunway University, Bandar Sunway, Malaysia
| | - Abubakr M. Idris
- Department of Chemistry, College of Science, King Khalid University, Abha, Saudi Arabia
- Research Center for Advanced Materials Science (RCAMS), King Khalid University, Abha, Saudi Arabia
- *Correspondence: Talha Bin Emran, ; Abubakr M. Idris, ; Bonglee Kim,
| | - Bonglee Kim
- Department of Pathology College of Korean Medicine, Kyung Hee University, Seoul, South Korea
- *Correspondence: Talha Bin Emran, ; Abubakr M. Idris, ; Bonglee Kim,
| |
Collapse
|
174
|
Xia HF, Lai WQ, Chen GH, Li Y, Xie QH, Jia YL, Chen G, Zhao YF. A histological study of vascular wall resident stem cells in venous malformations. Cell Tissue Res 2022; 390:229-243. [PMID: 35916917 DOI: 10.1007/s00441-022-03672-3] [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: 10/12/2021] [Accepted: 07/13/2022] [Indexed: 11/25/2022]
Abstract
Vascular wall resident stem cells (VW-SCs) play a key role in vascular formation and remodeling under both physiological and pathological situations. They not only serve as a reservoir to supply all types of vascular cells needed, but also regulate vascular homeostasis by paracrine effects. Venous malformations (VMs) are common congenital vascular malformations which are just characterized by the deficient quantity and abnormal function of vascular cells. However, the existence and role of VW-SCs in VMs is still unclear at present. In this study, the level and distribution of VW-SCs in 22 specimens of VMs were measured by immunochemistry, double-labeling immunofluorescence, and qPCR, followed by the Spearman rank correlation test. We found that both the protein and mRNA expression levels of CD34, vWF, VEGFR2, CD44, CD90, and CD105 were significantly downregulated in VMs compared with that in normal venules. VW-SCs were sporadically distributed or even absent within and outside the endothelium of VMs. The expression of the VW-SC-related markers was positively correlated with the density of both endothelial cells and perivascular cells. All those results and established evidence indicated that VW-SCs were more sporadically distributed with fewer amounts in VMs, which possibly contributing to the deficiency of vascular cells in VMs.
Collapse
Affiliation(s)
- Hou-Fu Xia
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.,Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Wen-Qiang Lai
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Gao-Hong Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Ye Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Qi-Hui Xie
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Yu-Lin Jia
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Gang Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China. .,Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
| | - Yi-Fang Zhao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China. .,Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
| |
Collapse
|
175
|
Yu R, Zhong J, Zhou Q, Ren W, Liu Z, Bian Y. Kaempferol prevents angiogenesis of rat intestinal microvascular endothelial cells induced by LPS and TNF-α via inhibiting VEGF/Akt/p38 signaling pathways and maintaining gut-vascular barrier integrity. Chem Biol Interact 2022; 366:110135. [DOI: 10.1016/j.cbi.2022.110135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 08/18/2022] [Accepted: 08/23/2022] [Indexed: 11/03/2022]
|
176
|
Tang KY, Heng JZX, Chai CHT, Chan CY, Low BQL, Chong SME, Loh HY, Li Z, Ye E, Loh XJ. Modified Bacterial Cellulose for Biomedical Applications. Chem Asian J 2022; 17:e202200598. [DOI: 10.1002/asia.202200598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/30/2022] [Indexed: 11/09/2022]
Affiliation(s)
- Karen Yuanting Tang
- Institute of Materials Research and Engineering Strategic Research Initiative 2 Fusionopolis Way, Innovis, #08-03 138634 Singapore SINGAPORE
| | - Jerry Zhi Xiong Heng
- Institute of Materials Research and Engineering Strategic Research Initiative 2 Fusionopolis Way, Innovis, #08-03 138634 Singapore SINGAPORE
| | - Casandra Hui Teng Chai
- Institute of Materials Research and Engineering Strategic Research Initiative 2 Fusionopolis Way, Innovis, #08-03 138634 Singapore SINGAPORE
| | - Chui Yu Chan
- Institute of Materials Research and Engineering Strategic Research Initiative 2 Fusionopolis Way, Innovis, #08-03 138634 Singapore SINGAPORE
| | - Beverly Qian Ling Low
- National University of Singapore Department of Materials Science and Engineering SINGAPORE
| | - Serene Ming En Chong
- Singapore Institute of Technology Food, Chemical and Biotechnology Cluster SINGAPORE
| | - Hong Yi Loh
- Nanyang Technological University Department of Materials Science and Engineering SINGAPORE
| | - Zibiao Li
- Institute of Materials Research and Engineering Strategic Research Initiative 2 Fusionopolis Way, Innovis, #08-03 138634 Singapore SINGAPORE
| | - Enyi Ye
- Institute of Materials Research and Engineering Strategic Research Initiative 2 Fusionopolis Way, Innovis, #8-03 138634 Singapore SINGAPORE
| | - Xian Jun Loh
- Institute of Materials Research and Engineering Strategic Research Initiative 2 Fusionopolis Way, Innovis, #08-03 138634 Singapore SINGAPORE
| |
Collapse
|
177
|
Jin X, Liu J, Wang W, Li J, Liu G, Qiu R, Yang M, Liu M, Yang L, Du X, Lei B. Identification of Age-associated Proteins and Functional Alterations in Human Retinal Pigment Epithelium. GENOMICS, PROTEOMICS & BIOINFORMATICS 2022; 20:633-647. [PMID: 35752290 PMCID: PMC9880895 DOI: 10.1016/j.gpb.2022.06.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/15/2022] [Accepted: 06/13/2022] [Indexed: 01/31/2023]
Abstract
Retinal pigment epithelium (RPE) has essential functions, such as nourishing and supporting the neural retina, and is of vital importance in the pathogenesis of age-related retinal degeneration. However, the exact molecular changes of RPE during aging remain poorly understood. Here, we isolated human primary RPE (hRPE) cells from 18 eye donors distributed over a wide age range (10-67 years old). A quantitative proteomic analysis was performed to analyze changes in their intracellular and secreted proteins. Age-group related subtypes and age-associated proteins were revealed and potential age-associated mechanisms were validated in ARPE-19 and hRPE cells. The results of proteomic data analysis and verifications suggest that RNF123- and RNF149-related protein ubiquitination plays an important role in protecting hRPE cells from oxidative damage during aging. In older hRPE cells, apoptotic signaling-related pathways were up-regulated, and endoplasmic reticulum organization was down-regulated both in the intracellular and secreted proteomes. Our work paints a detailed molecular picture of hRPE cells during the aging process and provides new insights into the molecular characteristics of RPE during aging and under other related clinical retinal conditions.
Collapse
Affiliation(s)
- Xiuxiu Jin
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou 450003, China,Branch of National Clinical Research Center for Ocular Disease, Henan Provincial People’s Hospital, Zhengzhou 450003, China,School of Medicine, Henan Provincial People’s Hospital, Henan University, Zhengzhou 450003, China
| | - Jingyang Liu
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou 450003, China,Branch of National Clinical Research Center for Ocular Disease, Henan Provincial People’s Hospital, Zhengzhou 450003, China
| | - Weiping Wang
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou 450003, China,Branch of National Clinical Research Center for Ocular Disease, Henan Provincial People’s Hospital, Zhengzhou 450003, China
| | - Jiangfeng Li
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Guangming Liu
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou 450003, China,Branch of National Clinical Research Center for Ocular Disease, Henan Provincial People’s Hospital, Zhengzhou 450003, China
| | - Ruiqi Qiu
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou 450003, China,Branch of National Clinical Research Center for Ocular Disease, Henan Provincial People’s Hospital, Zhengzhou 450003, China
| | - Mingzhu Yang
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou 450003, China,Branch of National Clinical Research Center for Ocular Disease, Henan Provincial People’s Hospital, Zhengzhou 450003, China
| | - Meng Liu
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou 450003, China,Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Lin Yang
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou 450003, China,Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaofeng Du
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou 450003, China,Branch of National Clinical Research Center for Ocular Disease, Henan Provincial People’s Hospital, Zhengzhou 450003, China
| | - Bo Lei
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou 450003, China,Branch of National Clinical Research Center for Ocular Disease, Henan Provincial People’s Hospital, Zhengzhou 450003, China,Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450001, China,Corresponding author.
| |
Collapse
|
178
|
Goswami AG, Basu S, Huda F, Pant J, Ghosh Kar A, Banerjee T, Shukla VK. An appraisal of vascular endothelial growth factor (VEGF): the dynamic molecule of wound healing and its current clinical applications. Growth Factors 2022; 40:73-88. [PMID: 35584274 DOI: 10.1080/08977194.2022.2074843] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Angiogenesis is a critical step of wound healing, and its failure leads to chronic wounds. The idea of restoring blood flow to the damaged tissues by promoting neo-angiogenesis is lucrative and has been researched extensively. Vascular endothelial growth factor (VEGF), a key dynamic molecule of angiogenesis has been investigated for its functions. In this review, we aim to appraise its biology, the comprehensive role of this dynamic molecule in the wound healing process, and how this knowledge has been translated in clinical application in various types of wounds. Although, most laboratory research on the use of VEGF is promising, its clinical applications have not met great expectations. We discuss various lacunae that might exist in making its clinical application unsuccessful for commercial use, and provide insight to the foundation for future research.
Collapse
Affiliation(s)
- Aakansha Giri Goswami
- Department of General surgery, All India Institute of Medical Sciences, Rishikesh, India
| | - Somprakas Basu
- Department of General surgery, All India Institute of Medical Sciences, Rishikesh, India
| | - Farhanul Huda
- Department of General surgery, All India Institute of Medical Sciences, Rishikesh, India
| | - Jayanti Pant
- Department of Physiology, All India Institute of Medical Sciences, Rishikesh, India
| | - Amrita Ghosh Kar
- Department of Pathology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Tuhina Banerjee
- Department of Microbiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Vijay Kumar Shukla
- Department of General Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
179
|
Potential of curcumin-loaded cubosomes for topical treatment of cervical cancer. J Colloid Interface Sci 2022; 620:419-430. [DOI: 10.1016/j.jcis.2022.04.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/29/2022] [Accepted: 04/05/2022] [Indexed: 12/14/2022]
|
180
|
Hachana S, Larrivée B. TGF-β Superfamily Signaling in the Eye: Implications for Ocular Pathologies. Cells 2022; 11:2336. [PMID: 35954181 PMCID: PMC9367584 DOI: 10.3390/cells11152336] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 02/06/2023] Open
Abstract
The TGF-β signaling pathway plays a crucial role in several key aspects of development and tissue homeostasis. TGF-β ligands and their mediators have been shown to be important regulators of ocular physiology and their dysregulation has been described in several eye pathologies. TGF-β signaling participates in regulating several key developmental processes in the eye, including angiogenesis and neurogenesis. Inadequate TGF-β signaling has been associated with defective angiogenesis, vascular barrier function, unfavorable inflammatory responses, and tissue fibrosis. In addition, experimental models of corneal neovascularization, diabetic retinopathy, proliferative vitreoretinopathy, glaucoma, or corneal injury suggest that aberrant TGF-β signaling may contribute to the pathological features of these conditions, showing the potential of modulating TGF-β signaling to treat eye diseases. This review highlights the key roles of TGF-β family members in ocular physiology and in eye diseases, and reviews approaches targeting the TGF-β signaling as potential treatment options.
Collapse
Affiliation(s)
- Soumaya Hachana
- Maisonneuve-Rosemont Hospital Research Center, Montreal, QC H1T 2M4, Canada
- Department of Ophthalmology, Université de Montréal, Montreal, QC H3C 3J7, Canada
| | - Bruno Larrivée
- Maisonneuve-Rosemont Hospital Research Center, Montreal, QC H1T 2M4, Canada
- Department of Ophthalmology, Université de Montréal, Montreal, QC H3C 3J7, Canada
| |
Collapse
|
181
|
Messeha SS, Zarmouh NO, Antonie L, Soliman KFA. Sanguinarine Inhibition of TNF-α-Induced CCL2, IKBKE/NF-κB/ERK1/2 Signaling Pathway, and Cell Migration in Human Triple-Negative Breast Cancer Cells. Int J Mol Sci 2022; 23:ijms23158329. [PMID: 35955463 PMCID: PMC9368383 DOI: 10.3390/ijms23158329] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/26/2022] [Accepted: 07/26/2022] [Indexed: 11/16/2022] Open
Abstract
Angiogenesis is a process that drives breast cancer (BC) progression and metastasis, which is linked to the altered inflammatory process, particularly in triple-negative breast cancer (TNBC). In targeting inflammatory angiogenesis, natural compounds are a promising option for managing BC. Thus, this study was designed to determine the natural alkaloid sanguinarine (SANG) potential for its antiangiogenic and antimetastatic properties in triple-negative breast cancer (TNBC) cells. The cytotoxic effect of SANG was examined in MDA-MB-231 and MDA-MB-468 cell models at a low molecular level. In this study, SANG remarkably inhibited the inflammatory mediator chemokine CCL2 in MDA-MB-231 and MDA-MB-468 cells. Furthermore, qRT-PCR confirmed with Western analysis studies showed that mRNA CCL2 repression was concurrent with reducing its main regulator IKBKE and NF-κB signaling pathway proteins in both TNBC cell lines. The total ERK1/2 protein was inhibited in the more responsive MDA-MB-231 cells. SANG exhibited a higher potential to inhibit cell migration in MDA-MB-231 cells compared to MDA-MB-468 cells. Data obtained in this study suggest a unique antiangiogenic and antimetastatic effect of SANG in the MDA-MB-231 cell model. These effects are related to the compound’s ability to inhibit the angiogenic CCL2 and impact the ERK1/2 pathway. Therefore, SANG use may be recommended as a component of the therapeutic strategy for TNBC.
Collapse
Affiliation(s)
- Samia S. Messeha
- Division of Pharmaceutical Sciences, College of Pharmacy & Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA; (S.S.M.); (L.A.)
| | - Najla O. Zarmouh
- Faculty of Medical Technology-Misrata, Libyan Ministry of Technical & Vocational Education, Misrata LY72, Libya;
| | - Lovely Antonie
- Division of Pharmaceutical Sciences, College of Pharmacy & Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA; (S.S.M.); (L.A.)
| | - Karam F. A. Soliman
- Division of Pharmaceutical Sciences, College of Pharmacy & Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA; (S.S.M.); (L.A.)
- Correspondence: ; Tel./Fax: +1-850-599-3306
| |
Collapse
|
182
|
Halogenated Flavonoid Derivatives Display Antiangiogenic Activity. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27154757. [PMID: 35897938 PMCID: PMC9331694 DOI: 10.3390/molecules27154757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/13/2022] [Accepted: 07/14/2022] [Indexed: 11/16/2022]
Abstract
Antiangiogenic agents attenuate tumours' growth and metastases and are therefore beneficial as an adjuvant or standalone cancer regimen. Drugs with dual antiproliferative and antiangiogenic activities can achieve anticancer efficacy and overcome acquired resistance. In this study, synthetic flavones (5a,b) with reported anticancer activity, and derivatives (4b and 6a), exhibited significant inhibition of endothelial cell tube formation (40-55%, 12 h) at 1 µM, which is comparable to sunitinib (50% inhibition at 1 µM, 48 h). Flavones (4b, 5a,b and 6a) also showed 25-37% reduction in HUVECs migration at 10 µM. In a Western blotting assay, 5a and 5b subdued VEGFR2 phosphorylation by 37% and 57%, respectively, suggesting that VEGFR2 may be their main antiangiogenic target. 5b displayed the best docking fit with VEGFR2 in an in silico study, followed by 5a, emphasizing the importance of the 7-hydroxyl group accompanied by a 4-C=S for activity. Conversely, derivatives with a 4-carbonyl moiety fitted poorly into the target's binding pocket, suggesting that their antiangiogenic activity depends on a different target. This study provides valuable insight into the Structure Activity Relationships (SAR) and modes of action of halogenated flavones with VEGFR2 and highlights their therapeutic potential as antiangiogenic/anticancer lead compounds.
Collapse
|
183
|
He L, Lu H, Chu J, Qin X, Gao J, Chen M, Weinstein LS, Yang J, Zhang Q, Zhang C, Zhang W. Endothelial G protein stimulatory α-subunit is a critical regulator of post-ischemic angiogenesis. Front Cardiovasc Med 2022; 9:941946. [PMID: 35958407 PMCID: PMC9358140 DOI: 10.3389/fcvm.2022.941946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/05/2022] [Indexed: 11/13/2022] Open
Abstract
Post-ischemic angiogenesis is a vital pathophysiological process in diseases such as peripheral arterial disease (PAD), heart ischemia, and diabetic retinopathy. The molecular mechanisms of post-ischemic angiogenesis are complicated and not fully elucidated. The G protein stimulatory alpha subunit (Gsα) is essential for hormone-stimulated cyclic adenosine monophosphate (cAMP) production and is an important regulator for many physiological processes. In the present study, we investigated the role of endothelial Gsα in post-ischemic angiogenesis by generating adult mice with endothelial-specific Gsα deficiency (GsαECKO). GsαECKO mice had impaired blood flow recovery after hind limb ischemic injury, and reduced neovascularization in allograft transplanted tumors. Mechanically, Gsα could regulate the expression of angiogenic factor with G patch and FHA domains 1 (AGGF1) through cAMP/CREB pathway. AGGF1 plays a key role in angiogenesis and regulates endothelial cell proliferation as well as migration. Knockdown of CREB or mutation of the CRE site on the AGGF1 promoter led to reduced AGGF1 promoter activity. In addition, knockdown of AGGF1 reduced the proangiogenic effect of Gsα in endothelial cells, and overexpression of AGGF1 reversed the impaired angiogenesis in GsαECKO mice in vivo. The finding may prove useful in designing new therapeutic targets for treatments of post-ischemic angiogenesis-related diseases.
Collapse
Affiliation(s)
- Lifan He
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hanlin Lu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Cardiovascular Disease Research Center of Shandong First Medical University, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jianying Chu
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, China
| | - Xiaoteng Qin
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jiangang Gao
- School of Life Sciences and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, China
| | - Min Chen
- Metabolic Diseases Branch, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Lee S. Weinstein
- Metabolic Diseases Branch, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Jianmin Yang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qunye Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Cheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wencheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Cardiovascular Disease Research Center of Shandong First Medical University, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
- *Correspondence: Wencheng Zhang,
| |
Collapse
|
184
|
Ai Y, Zhao Z, Wang H, Zhang X, Qin W, Guo Y, Zhao M, Tang J, Ma X, Zeng J. Pull the plug: Anti‐angiogenesis potential of natural products in gastrointestinal cancer therapy. Phytother Res 2022; 36:3371-3393. [PMID: 35871532 DOI: 10.1002/ptr.7492] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/13/2022] [Accepted: 04/28/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Yanling Ai
- Department of Oncology Hospital of Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Ziyi Zhao
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province Hospital of Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Hengyi Wang
- Department of Oncology Hospital of Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Xiaomei Zhang
- Institute of Medicinal Chemistry of Chinese Medicine Chongqing Academy of Chinese Materia Medica Chongqing China
| | - Weihan Qin
- Institute of Medicinal Chemistry of Chinese Medicine Chongqing Academy of Chinese Materia Medica Chongqing China
| | - Yanlei Guo
- Institute of Medicinal Chemistry of Chinese Medicine Chongqing Academy of Chinese Materia Medica Chongqing China
| | - Maoyuan Zhao
- Department of Oncology Hospital of Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Jianyuan Tang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province Hospital of Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy Chengdu University of Traditional Chinese Medicine Chengdu China
| | - Jinhao Zeng
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province Hospital of Chengdu University of Traditional Chinese Medicine Chengdu China
- Department of Geriatrics Hospital of Chengdu University of Traditional Chinese Medicine Chengdu China
| |
Collapse
|
185
|
Li J, Li R, Wu X, Zheng C, Shiu PHT, Rangsinth P, Lee SMY, Leung GPH. An Update on the Potential Application of Herbal Medicine in Promoting Angiogenesis. Front Pharmacol 2022; 13:928817. [PMID: 35928282 PMCID: PMC9345329 DOI: 10.3389/fphar.2022.928817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Angiogenesis, the formation of new capillaries from pre-existing vascular networks, plays an important role in many physiological and pathological processes. The use of pro-angiogenic agents has been proposed as an attractive approach for promoting wound healing and treating vascular insufficiency-related problems, such as ischemic heart disease and stroke, which are the leading causes of death worldwide. Traditional herbal medicine has a long history; however, there is still a need for more in-depth studies and evidence-based confirmation from controlled and validated trials. Many in vitro and in vivo studies have reported that herbal medicines and their bioactive ingredients exert pro-angiogenic activity. The most frequently studied pro-angiogenic phytochemicals include ginsenosides from Panax notoginseng, astragalosides and calycosin from Radix Astragali, salvianolic acid B from Salvia miltiorrhiza, paeoniflorin from Radix Paeoniae, ilexsaponin A1 from Ilex pubescens, ferulic acid from Angelica sinensis, and puerarin from Radix puerariae. This review summarizes the progress in research on these phytochemicals, particularly those related to pro-angiogenic mechanisms and applications in ischemic diseases, tissue repair, and wound healing. In addition, an outline of their limitations and challenges during drug development is presented.
Collapse
Affiliation(s)
- Jingjing Li
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Renkai Li
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Xiaoping Wu
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Chengwen Zheng
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Polly Ho-Ting Shiu
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Panthakarn Rangsinth
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Simon Ming-Yuen Lee
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Taipa Macao SAR, China
| | - George Pak-Heng Leung
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- *Correspondence: George Pak-Heng Leung,
| |
Collapse
|
186
|
Chen T, Jia F, Yu Y, Zhang W, Wang C, Zhu S, Zhang N, Liu X. Potential Role of Quercetin in Polycystic Ovary Syndrome and Its Complications: A Review. Molecules 2022; 27:molecules27144476. [PMID: 35889348 PMCID: PMC9325244 DOI: 10.3390/molecules27144476] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/08/2022] [Accepted: 07/08/2022] [Indexed: 12/18/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is a common multisystem disease with reproductive, metabolic and psychological abnormalities. It is characterized by a high prevalence rate in women of childbearing age and highly heterogeneous clinical manifestations, which seriously harm women’s physical and mental health. Quercetin (QUR) is a natural compound of flavonoids found in a variety of foods and medicinal plants. It can intervene with the pathologic process of PCOS from multiple targets and channels and has few adverse reactions. It is mentioned in this review that QUR can improve ovulation disorder, relieve Insulin resistance (IR), reduce androgen, regulate lipid metabolism, regulate gut microbiota and improve vascular endothelial function, which is of great significance in the treatment of PCOS.
Collapse
Affiliation(s)
- Tong Chen
- Department of Gynecology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; (T.C.); (F.J.); (Y.Y.); (W.Z.); (C.W.); (S.Z.); (N.Z.)
- China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Fan Jia
- Department of Gynecology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; (T.C.); (F.J.); (Y.Y.); (W.Z.); (C.W.); (S.Z.); (N.Z.)
- China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yue Yu
- Department of Gynecology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; (T.C.); (F.J.); (Y.Y.); (W.Z.); (C.W.); (S.Z.); (N.Z.)
- China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Wufan Zhang
- Department of Gynecology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; (T.C.); (F.J.); (Y.Y.); (W.Z.); (C.W.); (S.Z.); (N.Z.)
- China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Chaoying Wang
- Department of Gynecology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; (T.C.); (F.J.); (Y.Y.); (W.Z.); (C.W.); (S.Z.); (N.Z.)
- Department of Gynecology of Traditional Chinese Medicine, Graduate School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Shiqin Zhu
- Department of Gynecology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; (T.C.); (F.J.); (Y.Y.); (W.Z.); (C.W.); (S.Z.); (N.Z.)
- Department of Gynecology of Traditional Chinese Medicine, Graduate School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Nana Zhang
- Department of Gynecology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; (T.C.); (F.J.); (Y.Y.); (W.Z.); (C.W.); (S.Z.); (N.Z.)
- Department of Gynecology of Traditional Chinese Medicine, Graduate School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xinmin Liu
- Department of Gynecology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; (T.C.); (F.J.); (Y.Y.); (W.Z.); (C.W.); (S.Z.); (N.Z.)
- Correspondence:
| |
Collapse
|
187
|
Ma X, Wang J. Formononetin: A Pathway to Protect Neurons. Front Integr Neurosci 2022; 16:908378. [PMID: 35910340 PMCID: PMC9326316 DOI: 10.3389/fnint.2022.908378] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/14/2022] [Indexed: 11/19/2022] Open
Abstract
Formononetin (FMN) is a phytoestrogen member of the flavonoid family, which has the pharmacological effects of antioxidative, antihypertensive, antitumor, and anti-infective. FMN demonstrates potential in the prevention and treatment of diseases, specifically neurological diseases, such as traumatic brain injury (TBI), spinal cord injury (SCI), ischemic stroke, cerebral ischemia-reperfusion, Alzheimer’s disease, and nerve tumor. Herein, a literature search is conducted to provide information on the signaling pathways of neuroprotection of formononetin based on the neuroprotective study. The significant neuroprotective function of FMN makes it a novel candidate for the development of drugs targeting the central nervous system.
Collapse
Affiliation(s)
- Xiaoyu Ma
- The Second Clinical Medical School, Nanjing Medical University, Nanjing, China
| | - Juejin Wang
- Department of Physiology, Nanjing Medical University, Nanjing, China
- *Correspondence: Juejin Wang,
| |
Collapse
|
188
|
Ionescu C, Oprea B, Ciobanu G, Georgescu M, Bică R, Mateescu GO, Huseynova F, Barragan-Montero V. The Angiogenic Balance and Its Implications in Cancer and Cardiovascular Diseases: An Overview. Medicina (B Aires) 2022; 58:medicina58070903. [PMID: 35888622 PMCID: PMC9316440 DOI: 10.3390/medicina58070903] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 12/12/2022] Open
Abstract
Angiogenesis is the process of developing new blood vessels from pre-existing ones. This review summarizes the main features of physiological and pathological angiogenesis and those of angiogenesis activation and inhibition. In healthy adults, angiogenesis is absent apart from its involvement in female reproductive functions and tissue regeneration. Angiogenesis is a complex process regulated by the action of specific activators and inhibitors. In certain diseases, modulating the angiogenic balance can be a therapeutic route, either by inhibiting angiogenesis (for example in the case of tumor angiogenesis), or by trying to activate the process of new blood vessels formation, which is the goal in case of cardiac or peripheral ischemia.
Collapse
Affiliation(s)
- Cătălina Ionescu
- Department of Chemistry, Faculty of Sciences, University of Craiova, 107i Calea București, 200144 Craiova, Romania;
- Correspondence: (C.I.); (B.O.)
| | - Bogdan Oprea
- Histology Department, University of Medicine and Pharmacy, 2-4 Petru Rares, 200349 Craiova, Romania;
- Correspondence: (C.I.); (B.O.)
| | - Georgeta Ciobanu
- Department of Chemistry, Faculty of Sciences, University of Craiova, 107i Calea București, 200144 Craiova, Romania;
| | - Milena Georgescu
- Clinic for Plastic Surgery and Burns, County Emergency Hospital Craiova, 200642 Craiova, Romania;
| | - Ramona Bică
- General Hospital—“Victor Babes”, 281 Mihai Bravu St., Sector III, 030303 Bucharest, Romania;
| | - Garofiţa-Olivia Mateescu
- Histology Department, University of Medicine and Pharmacy, 2-4 Petru Rares, 200349 Craiova, Romania;
| | - Fidan Huseynova
- LBN, University of Montpellier, 34193 Montpellier, France; (F.H.); (V.B.-M.)
- Institute of Molecular Biology and Biotechnologies, Azerbaïjan National Academy of Sciences (ANAS), AZ1073 Baku, Azerbaijan
- Department of Histology, Cytology and Embryology, Azerbaijan Medical University, AZ1078 Baku, Azerbaijan
| | | |
Collapse
|
189
|
Wong AYW, Ong BSY, Lee ARYB, Mai AS, Selvarajan S, Lakshminarasappa SR, Tay SM. Topical Biological Agents as Adjuncts to Improve Wound Healing in Chronic Diabetic Wounds: A Systematic Review of Clinical Evidence and Future Directions. Cureus 2022; 14:e27180. [PMID: 36035037 PMCID: PMC9398533 DOI: 10.7759/cureus.27180] [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] [Accepted: 07/23/2022] [Indexed: 11/05/2022] Open
Abstract
Diabetes is a leading chronic illness in the modern world and 19-34% develop chronic diabetic foot ulcers (DFUs) in their lifetime, often necessitating amputation. The reduction in tissue growth factors and resulting imbalance between proteolytic enzymes and their inhibitors, along with systemic factors impairing healing appear particularly important in chronic wounds. Growth factors applied topically have thus been suggested to be a non-invasive, safe, and cost-effective adjunct to improve wound healing and prevent complications. Comprehensive database searches of MEDLINE via PubMed, EMBASE, Cochrane, and ClinicalTrials.gov were performed to identify clinical evidence and ongoing trials. The risk of bias analysis included randomized controlled trials (RCTs) was performed using the Cochrane Risk of Bias 2.0 tool. We included randomized controlled trials that compared the use of a topical biologic growth factor-containing regimen to any other regimen. Primary outcomes of interest were time to wound closure, healing rate, and time. Secondary outcomes included the incidence of adverse events such as infection. A total of 41 trials from 1992-2020 were included in this review, with a total recorded 3,112 patients. Platelet-derived growth factors (PDGF) in the form of becaplermin gel are likely to reduce the time of closure, increase the incidence of wound closure, and complete wound healing. Human umbilical cord-related treatments, dehydrated human amnion and chorion allograft (dHACA), and hypothermically stored amniotic membrane (HSAM), consistently increased the rates and incidence of complete ulcer healing while reducing ulcer size and time to complete ulcer healing. Fibroblast growth factor-1 (FGF1) showed only a slight benefit in multiple studies regarding increasing complete ulcer healing rates and incidence while reducing ulcer size and time to complete ulcer healing, with a few studies showing no statistical difference from placebo. Platelet-rich fibrin (PRF) is consistent in reducing the time to complete ulcer healing and increasing wound healing rate but may not reduce ulcer size or increase the incidence of complete ulcer healing. Targeting the wound healing pathway via the extrinsic administration of growth factors is a promising option to augment wound healing in diabetic patients. Growth factors have also shown promise in specific subgroups of patients who are at risk of significantly impaired wound healing such as those with a history of secondary infection and vasculopathy. As diabetes impairs multiple stages of wound healing, combining growth factors in diabetic wound care may prove to be an area of interest. Evidence from this systematic literature review suggests that topical adjuncts probably reduce time to wound closure, reduce healing time, and increase the healing rate in patients with chronic DFUs.
Collapse
Affiliation(s)
- Andrew Yew Wei Wong
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, SGP
| | | | | | - Aaron Shengting Mai
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, SGP
| | | | | | - Sook Muay Tay
- Surgical Intensive Care, Singapore General Hospital, Singapore, SGP
| |
Collapse
|
190
|
Angom RS, Kulkarni T, Wang E, Kumar Dutta S, Bhattacharya S, Das P, Mukhopadhyay D. Vascular Endothelial Growth Factor Receptor-1 Modulates Hypoxia-Mediated Endothelial Senescence and Cellular Membrane Stiffness via YAP-1 Pathways. Front Cell Dev Biol 2022; 10:903047. [PMID: 35846360 PMCID: PMC9283904 DOI: 10.3389/fcell.2022.903047] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 06/13/2022] [Indexed: 11/23/2022] Open
Abstract
Hypoxia-induced endothelial cell (EC) dysfunction has been implicated as potential initiators of different pathogenesis, including Alzheimer’s disease and vascular dementia. However, in-depth structural, mechanical, and molecular mechanisms leading to EC dysfunction and pathology need to be revealed. Here, we show that ECs exposed to hypoxic conditions readily enter a senescence phenotype. As expected, hypoxia upregulated the expression of vascular endothelial growth factor (VEGFs) and its receptors (VEGFRs) in the ECs. Interestingly, Knockdown of VEGFR-1 expression prior to hypoxia exposure prevented EC senescence, suggesting an important role of VEGFR-1 expression in the induction of EC senescence. Using atomic force microscopy, we showed that senescent ECs had a flattened cell morphology, decreased membrane ruffling, and increased membrane stiffness, demonstrating unique morphological and nanomechanical signatures. Furthermore, we show that hypoxia inhibited the Hippo pathway Yes-associated protein (YAP-1) expression and knockdown of YAP-1 induced senescence in the ECs, supporting a key role of YAP-1 expression in the induction of EC senescence. And importantly, VEGFR-1 Knockdown in the ECs modulated YAP-1 expression, suggesting a novel VEGFR-1-YAP-1 axis in the induction of hypoxia-mediated EC senescence. In conclusion, VEGFR-1 is overexpressed in ECs undergoing hypoxia-mediated senescence, and the knockdown of VEGFR-1 restores cellular structural and nanomechanical integrity by recovering YAP-1 expression.
Collapse
Affiliation(s)
| | - Tanmay Kulkarni
- Department of Biochemistry and Molecular Biology, Jacksonville, FL, United States
| | - Enfeng Wang
- Department of Biochemistry and Molecular Biology, Jacksonville, FL, United States
| | - Shamit Kumar Dutta
- Department of Biochemistry and Molecular Biology, Jacksonville, FL, United States
| | - Santanu Bhattacharya
- Department of Biochemistry and Molecular Biology, Jacksonville, FL, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Jacksonville, FL, United States
| | - Pritam Das
- Department of Biochemistry and Molecular Biology, Jacksonville, FL, United States
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Jacksonville, FL, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Jacksonville, FL, United States
- *Correspondence: Debabrata Mukhopadhyay,
| |
Collapse
|
191
|
Davenport BN, Wilson RL, Jones HN. Interventions for placental insufficiency and fetal growth restriction. Placenta 2022; 125:4-9. [PMID: 35414477 PMCID: PMC10947607 DOI: 10.1016/j.placenta.2022.03.127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/09/2022] [Accepted: 03/28/2022] [Indexed: 01/16/2023]
Abstract
Pregnancy complications adversely impact both mother and/or fetus throughout the lifespan. Fetal growth restriction (FGR) occurs when a fetus fails to reach their intrauterine potential for growth, it is the second highest leading cause of infant mortality, and leads to increased risk of developing non-communicable diseases in later life due 'fetal programming'. Abnormal placental development, growth and/or function underlies approximately 75% of FGR cases and there is currently no treatment save delivery, often prematurely. We previously demonstrated in a murine model of FGR that nanoparticle mediated, intra-placental human IGF-1 gene therapy maintains normal fetal growth. Multiple models of FGR currently exist reflecting the etiologies of human FGR and have been used by us and others to investigate the development of in utero therapeutics as discussed here. In addition to the in vivo models discussed herein, utilizing human models including in vitro (Choriocarcinoma cell lines and primary trophoblasts) and ex vivo (term villous fragments and placenta cotyledon perfusion) we have demonstrated robust nanoparticle uptake, transgene expression, nutrient transporter regulation without transfer to the fetus. For translational gene therapy application in the human placenta, there are multiple avenues that require investigation including syncytial uptake from the maternal circulation, transgene expression, functionality and longevity of treatment, impact of treatment on the mother and developing fetus. The potential impact of treating the placenta during gestation is high, wide-ranging across pregnancy complications, and may offer reduced risk of developing associated cardio-metabolic diseases in later life impacting at both an individual and societal level.
Collapse
Affiliation(s)
- Baylea N Davenport
- Center for Research in Perinatal Outcomes, University of Florida College of Medicine, United States
| | - Rebecca L Wilson
- Center for Research in Perinatal Outcomes, University of Florida College of Medicine, United States
| | - Helen N Jones
- Center for Research in Perinatal Outcomes, University of Florida College of Medicine, United States.
| |
Collapse
|
192
|
Sultan SR, Bashmail FT, Alzahrani NA, Alharbi SI, Anbar R, Alkharaiji M. Contrast-enhanced ultrasound for the evaluation of symptomatic and asymptomatic carotid plaques: A systematic review and meta-analysis. Echocardiography 2022; 39:1032-1043. [PMID: 35768892 DOI: 10.1111/echo.15407] [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: 02/14/2022] [Revised: 04/22/2022] [Accepted: 06/09/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Contrast-enhanced ultrasound (CEUS) is a promising imaging modality for the assessment of plaque vulnerability. We aimed to systematically review and meta-analyze the ability of CEUS parameters to differentiate between symptomatic and asymptomatic carotid plaques and to assess its reproducibility. METHODS PubMed, EMBASE, and Cochrane Library databases were searched for studies that potentially evaluated carotid plaques using CEUS. From the initial 2870 searches, 11 relevant publications comprising a total of 821 carotid plaques were reviewed. Data on CEUS parameters including quantitative and semi-quantitative parameters were extracted and analyzed. RESULTS The overall analysis showed significantly higher CEUS parameters in symptomatic carotid plaques compared to asymptomatic carotid plaques (standardised mean difference (SMD) .95, 95% confidence interval (CI) .56-1.27, p < .01). Intra and inter-observer reproducibility of quantitative CEUS parameters were excellent (intra-observer, r = .95, 95% CI .87-1; inter-observer, r = .93, 95% CI .80-.1). Semi-quantitative CEUS parameters showed good intra-observer reliability and moderate inter-observer reliability (intra-observer, r = .77, 95% CI .64-.89; inter-observer, r = .75, 95% CI .61-.89). Heterogeneity among studies compared CEUS parameters in symptomatic and asymptomatic plaques and studies assessed inter-observer reproducibility, and significant biases in studies assessing CEUS reproducibility were present. CONCLUSION CEUS is a useful vascular imaging method to differentiate between symptomatic and asymptomatic carotid plaques with moderate to excellent reproducibility. Quantitative CEUS analysis appeared to be more sensitive and reliable in assessing carotid plaques than semi-quantitative parameters. Further longitudinal prospective trials evaluating carotid plaque in asymptomatic population using CEUS to determine plaque characteristics that can become symptomatic are required.
Collapse
Affiliation(s)
- Salahaden R Sultan
- Department of Radiologic Sciences, College of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Fatima T Bashmail
- Department of Radiologic Sciences, College of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nouf A Alzahrani
- Department of Radiologic Sciences, College of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Shahd I Alharbi
- Department of Radiologic Sciences, College of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Rayan Anbar
- Department of Radiologic Sciences, College of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammed Alkharaiji
- Department of Public Health, College of Health Sciences, Saudi Electronic University, Riyadh, Saudi Arabia
| |
Collapse
|
193
|
He Y, Xu W, Xiao YT, Huang H, Gu D, Ren S. Targeting signaling pathways in prostate cancer: mechanisms and clinical trials. Signal Transduct Target Ther 2022; 7:198. [PMID: 35750683 PMCID: PMC9232569 DOI: 10.1038/s41392-022-01042-7] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 05/25/2022] [Accepted: 05/30/2022] [Indexed: 12/11/2022] Open
Abstract
Prostate cancer (PCa) affects millions of men globally. Due to advances in understanding genomic landscapes and biological functions, the treatment of PCa continues to improve. Recently, various new classes of agents, which include next-generation androgen receptor (AR) signaling inhibitors (abiraterone, enzalutamide, apalutamide, and darolutamide), bone-targeting agents (radium-223 chloride, zoledronic acid), and poly(ADP-ribose) polymerase (PARP) inhibitors (olaparib, rucaparib, and talazoparib) have been developed to treat PCa. Agents targeting other signaling pathways, including cyclin-dependent kinase (CDK)4/6, Ak strain transforming (AKT), wingless-type protein (WNT), and epigenetic marks, have successively entered clinical trials. Furthermore, prostate-specific membrane antigen (PSMA) targeting agents such as 177Lu-PSMA-617 are promising theranostics that could improve both diagnostic accuracy and therapeutic efficacy. Advanced clinical studies with immune checkpoint inhibitors (ICIs) have shown limited benefits in PCa, whereas subgroups of PCa with mismatch repair (MMR) or CDK12 inactivation may benefit from ICIs treatment. In this review, we summarized the targeted agents of PCa in clinical trials and their underlying mechanisms, and further discussed their limitations and future directions.
Collapse
Affiliation(s)
- Yundong He
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China.
| | - Weidong Xu
- Department of Urology, Shanghai Changzheng Hospital, Shanghai, China
| | - Yu-Tian Xiao
- Department of Urology, Shanghai Changzheng Hospital, Shanghai, China.,Department of Urology, Shanghai Changhai Hospital, Shanghai, China
| | - Haojie Huang
- Department of Urology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Di Gu
- Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Shancheng Ren
- Department of Urology, Shanghai Changzheng Hospital, Shanghai, China.
| |
Collapse
|
194
|
Wang F, Ding P, Liang X, Ding X, Brandt CB, Sjöstedt E, Zhu J, Bolund S, Zhang L, de Rooij LPMH, Luo L, Wei Y, Zhao W, Lv Z, Haskó J, Li R, Qin Q, Jia Y, Wu W, Yuan Y, Pu M, Wang H, Wu A, Xie L, Liu P, Chen F, Herold J, Kalucka J, Karlsson M, Zhang X, Helmig RB, Fagerberg L, Lindskog C, Pontén F, Uhlen M, Bolund L, Jessen N, Jiang H, Xu X, Yang H, Carmeliet P, Mulder J, Chen D, Lin L, Luo Y. Endothelial cell heterogeneity and microglia regulons revealed by a pig cell landscape at single-cell level. Nat Commun 2022; 13:3620. [PMID: 35750885 PMCID: PMC9232580 DOI: 10.1038/s41467-022-31388-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 06/16/2022] [Indexed: 11/23/2022] Open
Abstract
Pigs are valuable large animal models for biomedical and genetic research, but insights into the tissue- and cell-type-specific transcriptome and heterogeneity remain limited. By leveraging single-cell RNA sequencing, we generate a multiple-organ single-cell transcriptomic map containing over 200,000 pig cells from 20 tissues/organs. We comprehensively characterize the heterogeneity of cells in tissues and identify 234 cell clusters, representing 58 major cell types. In-depth integrative analysis of endothelial cells reveals a high degree of heterogeneity. We identify several functionally distinct endothelial cell phenotypes, including an endothelial to mesenchymal transition subtype in adipose tissues. Intercellular communication analysis predicts tissue- and cell type-specific crosstalk between endothelial cells and other cell types through the VEGF, PDGF, TGF-β, and BMP pathways. Regulon analysis of single-cell transcriptome of microglia in pig and 12 other species further identifies MEF2C as an evolutionally conserved regulon in the microglia. Our work describes the landscape of single-cell transcriptomes within diverse pig organs and identifies the heterogeneity of endothelial cells and evolutionally conserved regulon in microglia.
Collapse
Affiliation(s)
- Fei Wang
- Lars Bolund Institute of Regenerative Medicine, Qingdao-Europe Advanced Institute for Life Sciences, BGI-Qingdao, BGI-Shenzhen, Qingdao, China
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- BGI-Shenzhen, Shenzhen, China
| | - Peiwen Ding
- BGI-Shenzhen, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Xue Liang
- Lars Bolund Institute of Regenerative Medicine, Qingdao-Europe Advanced Institute for Life Sciences, BGI-Qingdao, BGI-Shenzhen, Qingdao, China
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Xiangning Ding
- BGI-Shenzhen, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Camilla Blunk Brandt
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Evelina Sjöstedt
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Jiacheng Zhu
- BGI-Shenzhen, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Saga Bolund
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Lijing Zhang
- BGI-Shenzhen, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- MGI, BGI-Shenzhen, Shenzhen, China
| | - Laura P M H de Rooij
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
- Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Lihua Luo
- BGI-Shenzhen, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yanan Wei
- BGI-Shenzhen, Shenzhen, China
- College of Basic Medicine, Qingdao University, Qingdao, China
| | - Wandong Zhao
- BGI-Shenzhen, Shenzhen, China
- College of Basic Medicine, Qingdao University, Qingdao, China
| | - Zhiyuan Lv
- BGI-Shenzhen, Shenzhen, China
- College of Basic Medicine, Qingdao University, Qingdao, China
| | - János Haskó
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Runchu Li
- BGI-Shenzhen, Shenzhen, China
- College of Basic Medicine, Qingdao University, Qingdao, China
| | - Qiuyu Qin
- BGI-Shenzhen, Shenzhen, China
- College of Basic Medicine, Qingdao University, Qingdao, China
| | - Yi Jia
- BGI-Shenzhen, Shenzhen, China
- College of Basic Medicine, Qingdao University, Qingdao, China
| | - Wendi Wu
- BGI-Shenzhen, Shenzhen, China
- College of Basic Medicine, Qingdao University, Qingdao, China
| | - Yuting Yuan
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, China
| | - Mingyi Pu
- BGI-Shenzhen, Shenzhen, China
- College of Basic Medicine, Qingdao University, Qingdao, China
| | - Haoyu Wang
- BGI-Shenzhen, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Aiping Wu
- Institute of Systems Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Suzhou Institute of Systems Medicine, Suzhou, China
| | - Lin Xie
- MGI, BGI-Shenzhen, Shenzhen, China
| | - Ping Liu
- MGI, BGI-Shenzhen, Shenzhen, China
| | | | | | - Joanna Kalucka
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Aarhus University of Advanced Studies (AIAS), Aarhus University, Aarhus, Denmark
| | - Max Karlsson
- Department of Protein Science, Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Xiuqing Zhang
- BGI-Shenzhen, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Rikke Bek Helmig
- Department of Obstetrics and Gynecology, Aarhus University Hospital, Aarhus, Denmark
| | - Linn Fagerberg
- Department of Protein Science, Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Cecilia Lindskog
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Fredrik Pontén
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Mathias Uhlen
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Protein Science, Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Lars Bolund
- Lars Bolund Institute of Regenerative Medicine, Qingdao-Europe Advanced Institute for Life Sciences, BGI-Qingdao, BGI-Shenzhen, Qingdao, China
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Niels Jessen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | | | - Xun Xu
- BGI-Shenzhen, Shenzhen, China
| | - Huanming Yang
- BGI-Shenzhen, Shenzhen, China
- IBMC-BGI Center, the Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Peter Carmeliet
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
- Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Jan Mulder
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Dongsheng Chen
- BGI-Shenzhen, Shenzhen, China.
- Institute of Systems Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.
- Suzhou Institute of Systems Medicine, Suzhou, China.
| | - Lin Lin
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark.
| | - Yonglun Luo
- Lars Bolund Institute of Regenerative Medicine, Qingdao-Europe Advanced Institute for Life Sciences, BGI-Qingdao, BGI-Shenzhen, Qingdao, China.
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
- BGI-Shenzhen, Shenzhen, China.
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark.
- IBMC-BGI Center, the Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China.
| |
Collapse
|
195
|
Cissy Yu Q, Bai L, Chen Y, Chen Y, Peng G, Wang D, Yang G, Cui G, Jing N, Arial Zeng Y. Embryonic vascular establishment requires protein C receptor-expressing endothelial progenitors. Development 2022; 149:275466. [DOI: 10.1242/dev.200419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 05/05/2022] [Indexed: 11/20/2022]
Abstract
ABSTRACT
Vascular establishment is one of the early events in embryogenesis. It is believed that vessel-initiating endothelial progenitors cluster to form the first primitive vessel. Understanding the molecular identity of these progenitors is crucial in order to elucidate lineage hierarchy. In this study, we identify protein C receptor (Procr) as an endothelial progenitor marker and investigate the role of Procr+ progenitors during embryonic vascular development. Using a ProcrmGFP-2A-lacZ reporter, we reveal a much earlier Procr expression (embryonic day 7.5) than previously acknowledged (embryonic day 13.5). Genetic fate-mapping experiments using ProcrCre and ProcrCreER demonstrate that Procr+ cells give rise to blood vessels throughout the entire embryo proper. Single-cell RNA-sequencing analyses place Procr+ cells at the start of endothelial commitment and maturation. Furthermore, targeted ablation of Procr+ cells results in failure of vessel formation and early embryonic lethality. Notably, genetic fate mapping and scRNA-seq pseudotime analysis support the view that Procr+ progenitors can give rise to hemogenic endothelium. In this study, we establish a Procr expression timeline and identify Procr+ vessel-initiating progenitors, and demonstrate their indispensable role in establishment of the vasculature during embryo development.
Collapse
Affiliation(s)
- Qing Cissy Yu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences 1 , Shanghai 200031 , China
| | - Lanyue Bai
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences 1 , Shanghai 200031 , China
| | - Yingying Chen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences 1 , Shanghai 200031 , China
| | - Yujie Chen
- CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, CAS 3 Key Laboratory of Computational Biology , , Shanghai 200031 , China
| | - Guangdun Peng
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong Institutes of Biomedicine and Health, Chinese Academy of Sciences 4 , Guangzhou 510530 , China
| | - Daisong Wang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences 1 , Shanghai 200031 , China
| | - Guowei Yang
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences 2 , 310024 Hangzhou , China
| | - Guizhong Cui
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong Institutes of Biomedicine and Health, Chinese Academy of Sciences 4 , Guangzhou 510530 , China
| | - Naihe Jing
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences 1 , Shanghai 200031 , China
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong Institutes of Biomedicine and Health, Chinese Academy of Sciences 4 , Guangzhou 510530 , China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences 5 , Beijing 100101 , China
| | - Yi Arial Zeng
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences 1 , Shanghai 200031 , China
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences 2 , 310024 Hangzhou , China
| |
Collapse
|
196
|
Obesity: The Fat Tissue Disease Version of Cancer. Cells 2022; 11:cells11121872. [PMID: 35741001 PMCID: PMC9221301 DOI: 10.3390/cells11121872] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 06/02/2022] [Accepted: 06/06/2022] [Indexed: 02/06/2023] Open
Abstract
Obesity is a disease with high potential for fatality. It perfectly fits the disease definition, as cancer does. This is because it damages body structure and functions, both mechanically and biologically, and alters physical, mental, and social health. In addition, it shares many common morbid characteristics with the most feared disease, cancer. For example, it is influenced by a sophisticated interaction between a person’s genetics, the environment, and an increasing number of other backgrounds. Furthermore, it displays abnormal cell growth and proliferation events, only limited to white fat, resulting in adipose tissue taking up an increasing amount of space within the body. This occurs through fat “metastases” and via altered signaling that further aggravates the pathology of obesity by inducing ubiquitous dishomeostasis. These metastases can be made graver by angiogenesis, which might boost diseased tissue growth. More common features with cancer include its progressive escalation through different levels of severity and its possibility of re-onset after recovery. Despite all these similarities with cancer, obesity is substantially less agitating for most people. Thus, the ideas proposed herein could have utility to sensitize the public opinion about the hard reality of obesity. This is increasingly needed, as the obesity pandemic has waged a fierce war against our bodies and society in general, while there is still doubt about whether it is a real disease or not. Hence, raising public consciousness to properly face health issues is crucial to improving our health instead of gaining weight unhealthily. It is obviously illogical to fight cancer extremely seriously on the one hand and to consider dying with obesity as self-inflicted on the other. In fact, obesity merits a top position among the most lethal diseases besides cancer.
Collapse
|
197
|
Akbarian M, Bertassoni LE, Tayebi L. Biological aspects in controlling angiogenesis: current progress. Cell Mol Life Sci 2022; 79:349. [PMID: 35672585 PMCID: PMC10171722 DOI: 10.1007/s00018-022-04348-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/01/2022] [Accepted: 05/03/2022] [Indexed: 12/25/2022]
Abstract
All living beings continue their life by receiving energy and by excreting waste products. In animals, the arteries are the pathways of these transfers to the cells. Angiogenesis, the formation of the arteries by the development of pre-existed parental blood vessels, is a phenomenon that occurs naturally during puberty due to certain physiological processes such as menstruation, wound healing, or the adaptation of athletes' bodies during exercise. Nonetheless, the same life-giving process also occurs frequently in some patients and, conversely, occurs slowly in some physiological problems, such as cancer and diabetes, so inhibiting angiogenesis has been considered to be one of the important strategies to fight these diseases. Accordingly, in tissue engineering and regenerative medicine, the highly controlled process of angiogenesis is very important in tissue repairing. Excessive angiogenesis can promote tumor progression and lack of enough angiogensis can hinder tissue repair. Thereby, both excessive and deficient angiogenesis can be problematic, this review article introduces and describes the types of factors involved in controlling angiogenesis. Considering all of the existing strategies, we will try to lay out the latest knowledge that deals with stimulating/inhibiting the angiogenesis. At the end of the article, owing to the early-reviewed mechanical aspects that overshadow angiogenesis, the strategies of angiogenesis in tissue engineering will be discussed.
Collapse
Affiliation(s)
- Mohsen Akbarian
- Department of Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
| | - Luiz E Bertassoni
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, OR, USA
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI, 53233, USA.
| |
Collapse
|
198
|
Fang C, Zuo K, Wang H, Zhang W, Zhong J, Xu L, Yang X. Association of plasma Ninjurin-1 and SYNTAX score in patients with coronary artery disease. Clin Biochem 2022; 108:50-55. [DOI: 10.1016/j.clinbiochem.2022.06.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 05/31/2022] [Accepted: 06/23/2022] [Indexed: 11/30/2022]
|
199
|
Ren A, Gan Q, Han W, Gong D, Cai J, Qu X. Endothelial GATA5 positively regulates angiogenesis via cathepsin S-mediated Angpt2/Flk1 and MMP2/9 signaling pathways. Biochem Biophys Res Commun 2022; 609:111-118. [DOI: 10.1016/j.bbrc.2022.04.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/03/2022] [Accepted: 04/05/2022] [Indexed: 12/24/2022]
|
200
|
Tsivelekas KK, Evangelopoulos DS, Pallis D, Benetos IS, Papadakis SA, Vlamis J, Pneumaticos SG. Angiogenesis in Spinal Cord Injury: Progress and Treatment. Cureus 2022; 14:e25475. [PMID: 35800787 PMCID: PMC9246426 DOI: 10.7759/cureus.25475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/29/2022] [Indexed: 11/22/2022] Open
Abstract
Traumatic spinal cord injury (SCI) provokes the onset of an intricate pathological process. Initial primary injury ruptures local micro-neuro-vascularcomplex triggering the commencement of multi-factorial secondary sequences which exert significant influence on neurological deterioration progress. Stimulating by local ischemia, neovascularization pathways emerge to provide neuroprotection and improve functional recovery. Although angiogenetic processes are prompted, newly formed vascular system is frequently inadequate to distribute sufficient blood supply and improve axonal recovery. Several treatment interventions have been endeavored to achieve the optimal conditions in SCI microenvironment, enhancing angiogenesis and improve functional recovery. In this study we review the revascularization pathogenesis and importance within the secondary processes and condense the proangiogenic influence of several angiogenetic-targeted treatment interventions.
Collapse
|