1
|
Allahyari Z, Lotfabadi NN, Zare F. The role of leukemia inhibitory factor in regulating angiogenesis-related gene expression in a mouse model of recurrent miscarriage. Placenta 2025; 165:91-101. [PMID: 40228388 DOI: 10.1016/j.placenta.2025.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 04/03/2025] [Accepted: 04/08/2025] [Indexed: 04/16/2025]
Abstract
BACKGROUND Recurrent miscarriage is an early pregnancy complication that affects approximately 1-3 % of pregnant couples. Leukemia Inhibitory Factor (LIF) plays an important role in various biological processes, including angiogenesis and pregnancy. This study aimed to evaluate the role of LIF in regulating angiogenesis-related genes in a mouse model of recurrent miscarriage. METHOD Female CBA/J mice mated with DBA/2J males were utilized as a miscarriage model. The study population was randomly assigned to three groups, normal group, mating female CBA/J mouse with male Balb/c without injection; miscarriage model control group with PBS injection; and the miscarriage group, in which LIF was injected. Following detection of a vaginal plug, mice were dissected on days 4, 7, and 14 of pregnancy. Uterine and placental tissues were collected to assess the expression of angiogenesis-related genes, including VEGF, PDGF, ANG1, FGF, and TGF-β, using real-time PCR. RESULT Data analysis revealed no significant differences in the expression of angiogenesis-related genes on days 4 and 7 of pregnancy compared with the control group. However, on day 14 of pregnancy, the expression of VEGF and TGF-β was significantly elevated in the miscarriage group receiving LIF compared to other groups (P = 0.03 and P = 0.04, respectively). The placental expression of the studied genes also exhibited a non-significant increase in the miscarriage group, with VEGF and TGF-β showing the most prominent increases, although these changes were not statistically significant. Correlation analysis between uterine and placental gene expression on day 14 revealed no significant association. CONCLUSION LIF regulates the uterine and placental expression of angiogenesis-related genes, particularly VEGF and TGF-β. These findings highlight the role of LIF in regulating angiogenesis-related gene expression and suggest that LIF could be a potential therapeutic candidate for improving pregnancy outcomes in cases of recurrent miscarriage.
Collapse
Affiliation(s)
- Zahra Allahyari
- Molecular Genetics, Faculty of Science, Science and Arts University, Yazd, Iran
| | | | - Fateme Zare
- Reproductive Immunology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
2
|
Ben Hassine A, Petit C, Thomas M, Mundweiler S, Guignandon A, Avril S. Gene expression modulation in human aortic smooth muscle cells under induced physiological mechanical stretch. Sci Rep 2024; 14:31147. [PMID: 39732782 DOI: 10.1038/s41598-024-82495-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 12/05/2024] [Indexed: 12/30/2024] Open
Abstract
In this study, we investigated gene expression in vitro of human primary Aortic smooth muscle cells (AoSMCs) in response to 9% physiological dynamic stretch over a 4 to 72-h timeframe using RT-qPCR. AoSMC were derived from primary culture and were exposed to continuous cycles of stretch and relaxation at 1 Hz by a computer-controlled Flex Jr.™ Tension System. Unstretched control AoSMCs were simultaneously cultured in the same dishes. Our results revealed a rapid and significant upregulation of specific genes (COL1A1, FBN1, LAMA5, TGFBR1 and TGFBR2) within the initial 4 h for AoSMCs subjected to dynamic stretching, whilst control cells did not respond within the same 4 h. The upregulated genes were the ones associated with extracellular matrix (ECM) fibrillogenesis and regulation of traction forces. Interestingly, stretched cells maintained stable gene expression between 4 and 72 h, whilst control cells exhibited variations over time in the absence of mechanical cues. These findings shed light on the essential role played by pulsatile stretches in the regulation of gene expressions by AoSMCs and the intricate processes governing their mechanobiological function, paving the way for further investigations in cardiovascular health.
Collapse
Affiliation(s)
- Amira Ben Hassine
- Mines Saint-Etienne, Université Jean Monnet, INSERM, U 1059 SAINBIOSE, Saint-Etienne, 42023, France
| | - Claudie Petit
- Mines Saint-Etienne, Université Jean Monnet, INSERM, U 1059 SAINBIOSE, Saint-Etienne, 42023, France
| | - Mireille Thomas
- Mines Saint-Etienne, Université Jean Monnet, INSERM, U 1059 SAINBIOSE, Saint-Etienne, 42023, France
| | - Stéphanie Mundweiler
- Mines Saint-Etienne, Université Jean Monnet, INSERM, U 1059 SAINBIOSE, Saint-Etienne, 42023, France
| | - Alain Guignandon
- Mines Saint-Etienne, Université Jean Monnet, INSERM, U 1059 SAINBIOSE, Saint-Etienne, 42023, France
| | - Stéphane Avril
- Mines Saint-Etienne, Université Jean Monnet, INSERM, U 1059 SAINBIOSE, Saint-Etienne, 42023, France.
| |
Collapse
|
3
|
Yin QZ, Liu YJ, Zhang Q, Xi SY, Yang TB, Li JP, Gao J. Overexpression of Basonuclin Zinc Finger Protein 2 in stromal cell is related to mesenchymal phenotype and immunosuppression of mucinous colorectal adenocarcinoma. Int Immunopharmacol 2024; 142:113184. [PMID: 39306894 DOI: 10.1016/j.intimp.2024.113184] [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: 07/02/2024] [Revised: 09/02/2024] [Accepted: 09/13/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Mucinous carcinoma (MC) is a distinct histologic subtype of colorectal cancer (CRC) that is less studied and associated with poor prognosis. This study aimed to identify MC-specific therapeutic targets and biomarkers to improve the prognosis of this aggressive disease. METHODS CRC samples from The Cancer Genome Atlas (TCGA) were categorized into MC and non-MC (NMC) groups based on histologic type. A multi-scale embedded gene co-expression network analysis (MEGENA) was constructed to identify gene modules associated with the MC group. The potential functions of Basonuclin Zinc Finger Protein 2 (BNC2) were further analyzed using the Biomarker Exploration for Solid Tumors (BEST) database. In vivo and in vitro experiments were conducted to validate the predicted results. RESULTS We identified the stromal component-related gene, BNC2, in the MC population. This gene is associated with a shorter progression-free interval (PFI) in CRC patients. BNC2 promotes FAP (encoding Fibroblast Activation Protein Alpha) transcription in cancer-associated fibroblasts (CAFs) and is involved in angiogenesis through two pathways. Additionally, BNC2 enhances tumor cell invasiveness in a CAF-dependent manner. Patients with high BNC2 expression benefited less from immunotherapy compared to those with low BNC2 expression. CONCLUSIONS Our study highlights the clinical importance of BNC2 in MC, and targeting BNC2 on stromal cells (fibroblasts and endothelial cells) may be an effective strategy for treating MC.
Collapse
Affiliation(s)
- Qing-Zhong Yin
- Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Yuan-Jie Liu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, China
| | - Qian Zhang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, China
| | - Song-Yang Xi
- Zhenjiang Hospital of Chinese Traditional and Western Medicine, Zhenjiang, Jiangsu 212000, China
| | - Tian-Bao Yang
- Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Jie-Pin Li
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, China.
| | - Ju Gao
- The Yangzhou Clinical Medical College of Xuzhou Medical University, Yangzhou, Jiangsu 225009, China; Northern Jiangsu People's Hospital, Yangzhou, Jiangsu 225009, China.
| |
Collapse
|
4
|
Nazzal MK, Battina HL, Tewari NP, Mostardo SL, Nagaraj RU, Zhou D, Awosanya OD, Majety SK, Samson S, Blosser RJ, Dadwal UC, Mulcrone PL, Kacena MA. The effects of young and aged, male and female megakaryocyte conditioned media on angiogenic properties of endothelial cells. Aging (Albany NY) 2024; 16:13181-13200. [PMID: 39578050 PMCID: PMC11719103 DOI: 10.18632/aging.206077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 07/11/2024] [Indexed: 11/24/2024]
Abstract
With aging, the risk of fractures and compromised healing increases. Angiogenesis plays a significant role in bone healing and is impaired with aging. We have previously shown the impact of megakaryocytes (MKs) in regulating bone healing. Notably, MKs produce factors known to promote angiogenesis. We examined the effects of conditioned media (CM) generated from MKs derived from young (3-4-month-old) and aged (22-24-month-old), male and female C57BL/6J mice on bone marrow endothelial cell (BMEC) growth and function. Female MK CM, regardless of age, caused a >65% increase in BMEC proliferation and improved vessel formation by >115%. Likewise, young male MK CM increased vessel formation by 160%. Although aged male MK CM resulted in >150% increases in the formation of vascular nodes and meshes, 62% fewer vessels formed compared to young male MK CM treatment. Aged female MK CM improved migration by over 2500%. However, aged female and male MK CM caused less wound closure. MK CM treatments also significantly altered the expression of several genes including PDGFRβ, CXCR4, and CD36 relative to controls and between ages. Further testing of mechanisms responsible for age-associated differences may allow for novel strategies to improve MK-mediated angiogenesis and bone healing, particularly within the aging population.
Collapse
Affiliation(s)
- Murad K. Nazzal
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Hanisha L. Battina
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Nikhil P. Tewari
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sarah L. Mostardo
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Richard L. Roudebush Veterans' Administration Medical Center, Indianapolis, IN 46202, USA
| | - Rohit U. Nagaraj
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Donghui Zhou
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Olatundun D. Awosanya
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Saveda K. Majety
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sue Samson
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Rachel J. Blosser
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Richard L. Roudebush Veterans' Administration Medical Center, Indianapolis, IN 46202, USA
| | - Ushashi C. Dadwal
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Richard L. Roudebush Veterans' Administration Medical Center, Indianapolis, IN 46202, USA
| | - Patrick L. Mulcrone
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Richard L. Roudebush Veterans' Administration Medical Center, Indianapolis, IN 46202, USA
| | - Melissa A. Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Richard L. Roudebush Veterans' Administration Medical Center, Indianapolis, IN 46202, USA
| |
Collapse
|
5
|
Atiakshin D, Nikolaeva E, Semyachkina A, Kostin A, Volodkin A, Morozov S, Ignatyuk M, Mikhaleva L, Demyashkin G, Elieh-Ali-Komi D, Buchwalow I, Tiemann M. The Contribution of Mast Cells to the Regulation of Elastic Fiber Tensometry in the Skin Dermis of Children with Marfan Syndrome. Int J Mol Sci 2024; 25:9191. [PMID: 39273142 PMCID: PMC11394836 DOI: 10.3390/ijms25179191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/15/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024] Open
Abstract
Marfan syndrome (MFS) is a hereditary condition accompanied by disorders in the structural and regulatory properties of connective tissue, including elastic fibers, due to a mutation in the gene encodes for fibrillin-1 protein (FBN1 gene) and the synthesis of abnormal fibrillin-1 glycoprotein. Despite the high potential of mast cells (MCs) to remodel the extracellular matrix (ECM), their pathogenetic significance in MFS has not been considered yet. The group of patients with Marfan syndrome included two mothers and five children (three girls aged 4, 11, and 11 and two boys aged 12 and 13). Normal skin was examined in two children aged 11 and 12. Histochemical, monoplex, and multiplex immunohistochemical techniques; combined protocols of simultaneous histochemical and immunohistochemical staining (the results of staining were assessed using light, epifluorescence, and confocal microscopy); and bioinformatics algorithms for the quantitative analysis of detected targets were used to evaluate mast cells and their relationship with other cells from extracellular structures in the skin dermis. Analysis of the skin MC population in children with Marfan syndrome revealed a considerably increased number of intra-organic populations with the preservation of the specific Tryptase+Chymase+CPA3+ protease profile typical of the skin. The features of the MC histotopography phenotype in MFS consisted of closer colocalization with elastic fibers, smooth muscle cells, and fibroblasts. MCs formed many intradermal clusters that synchronized the activity of cell functions in the stromal landscape of the tissue microenvironment with the help of spatial architectonics, including the formation of cell chains and the creation of fibrous niches. In MCs, the expression of specific proteases, TGF-β, and heparin increased, with targeted secretion of biologically active substances relative to the dermal elastic fibers, which had specific structural features in MFS, including abnormal variability in thickness along their entire length, alternating thickened and thinned areas, and uneven surface topography. This paper discusses the potential role of MCs in strain analysis (tensometry) of the tissue microenvironment in MFS. Thus, the quantitative and qualitative rearrangements of the skin MC population in MFS are aimed at altering the stromal landscape of the connective tissue. The results obtained should be taken into account when managing clinical signs of MFS manifested in other pathogenetically critical structures of internal organs, including the aorta, tendons, cartilage, and parenchymal organs.
Collapse
Affiliation(s)
- Dmitrii Atiakshin
- RUDN University, 6 Miklukho-Maklaya St., 117198 Moscow, Russian Federation
- Research Institute of Experimental Biology and Medicine, Burdenko Voronezh State Medical University, 394036 Voronezh, Russia
| | - Ekaterina Nikolaeva
- Veltischev Research and Clinical Institute for Pediatrics & Pediatric Surgery of the Pirogov Russian National Research Medical University, 2, Taldomskaya St., 125412 Moscow, Russia
| | - Alla Semyachkina
- Veltischev Research and Clinical Institute for Pediatrics & Pediatric Surgery of the Pirogov Russian National Research Medical University, 2, Taldomskaya St., 125412 Moscow, Russia
| | - Andrey Kostin
- RUDN University, 6 Miklukho-Maklaya St., 117198 Moscow, Russian Federation
| | - Artem Volodkin
- RUDN University, 6 Miklukho-Maklaya St., 117198 Moscow, Russian Federation
| | - Sergey Morozov
- Veltischev Research and Clinical Institute for Pediatrics & Pediatric Surgery of the Pirogov Russian National Research Medical University, 2, Taldomskaya St., 125412 Moscow, Russia
| | - Michael Ignatyuk
- RUDN University, 6 Miklukho-Maklaya St., 117198 Moscow, Russian Federation
| | - Liudmila Mikhaleva
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Center of Surgery, 119435 Moscow, Russia
| | - Grigory Demyashkin
- Laboratory of Histology and Immunohistochemistry, I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya St., 8/2, 119048 Moscow, Russia
| | - Daniel Elieh-Ali-Komi
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Allergology and Immunology, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 12203 Berlin, Germany
| | - Igor Buchwalow
- RUDN University, 6 Miklukho-Maklaya St., 117198 Moscow, Russian Federation
- Institute for Hematopathology, Fangdieckstr, 75a, 22547 Hamburg, Germany
| | - Markus Tiemann
- Institute for Hematopathology, Fangdieckstr, 75a, 22547 Hamburg, Germany
| |
Collapse
|
6
|
Hosseini E, Ahmadi J, Kargar F, Ghasemzadeh M. Coronary artery bypass grafting (CABG) induces pro-inflammatory and immunomodulatory phenotype of platelets in the absence of a pro-aggregatory state. Microvasc Res 2024; 153:104669. [PMID: 38360131 DOI: 10.1016/j.mvr.2024.104669] [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: 12/31/2023] [Revised: 02/06/2024] [Accepted: 02/08/2024] [Indexed: 02/17/2024]
Abstract
BACKGROUND Coronary artery bypass grafting (CABG) is considered the choice treatment for patients suffering from coronary artery disease (CAD). In the inflammatory milieu of cardiopulmonary bypass (CPB), systemic inflammatory response syndrome (SIRS) can induce a platelet pro-inflammatory state which could exacerbate post-CABG inflammatory status while affecting hemostatic function in patients. Therefore, focusing on platelets, the study presented here attempted to evaluate the pro-inflammatory and immunomodulatory profile of platelets as well as pro-aggregatory status during CABG. METHODS Platelets from patients undergoing CABG were subjected to flowcytometry analysis to evaluate P-selectin and CD40L expressions and PAC-1 binding in five intervals of 24 h before surgery, immediately, 2 h, 24 h, and one week after surgery. Moreover, intra-platelet TGF-β1 was also examined with western blotting. RESULTS Data showed increases of P-selectin and CD40L expressions in patients, with the meaningful loss of platelet contents of TGF-β1 after CABG (p < 0.001), where the changes tended to recover by day 7 of surgery while remaining above baseline (p < 0.001). Meanwhile, no significant change in PAC-1 binding capacity was shown. CONCLUSION The study presented here suggests that although the release of pro-inflammatory substances from platelets during CABG supports the post-operative inflammatory state, platelets are not pro-aggregatory enough to enhance thrombotic events after surgery. Whilst these observations could be due to successful medical interventions to optimize hemostasis during and after surgery, post-CABG reversal of anticoagulant by protamine is considered as another factor that may also have contributed to preventing pro-aggregatory but not pro-inflammatory and immunomodulatory functions of platelets.
Collapse
Affiliation(s)
- Ehteramolsadat Hosseini
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Javad Ahmadi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran; Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Faranak Kargar
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Science, Tehran, Iran.
| | - Mehran Ghasemzadeh
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran.
| |
Collapse
|
7
|
Guo Y, Wang Z, Zhou H, Pan H, Han W, Deng Y, Li Q, Xue J, Ge X, Wang S, Wang J, Zhang Y, Zhao C, Zhu H, Wang Y, Shen H, Liu D, Li J. First-in-human study of GFH018, a small molecule inhibitor of transforming growth factor-β receptor I inhibitor, in patients with advanced solid tumors. BMC Cancer 2024; 24:444. [PMID: 38600507 PMCID: PMC11007962 DOI: 10.1186/s12885-024-12216-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 04/02/2024] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND Transforming growth factor-β (TGF-β) is a cytokine with multiple functions, including cell growth regulation, extracellular matrix production, angiogenesis homeostasis adjustment and et al. TGF-β pathway activation promotes tumor metastasis/progression and mediates epithelial-mesenchymal transmission suppressing immunosurveillance in advanced tumors. GFH018, a small molecule inhibitor blocking TGF-β signal transduction, inhibits the progression and/or metastasis of advanced cancers. This first-in-human study evaluated the safety, tolerability, pharmacokinetics (PK), and efficacy of GFH018 monotherapy in patients with advanced solid tumors. METHODS This phase I, open-label, multicenter study used a modified 3+3 dose escalation and expansion design. Adult patients with advanced solid tumors failing the standard of care were enrolled. Starting at 5 mg, eight dose levels up to 85 mg were evaluated. Patients received GFH018 BID (14d-on/14d-off) starting on the 4th day after a single dose on cycle 1, day 1. Subsequent cycles were defined as 28 days. The study also explored the safety of 85 mg BID 7d-on/7d-off. Adverse events were graded using NCI criteria for adverse events (NCI-CTCAE v5.0). PK was analyzed using a noncompartmental method. Efficacy was evaluated using RECIST 1.1. Blood samples were collected for biomarker analysis. RESULTS Fifty patients were enrolled and received at least one dose of GFH018. No dose-limiting toxicity occurred, and the maximum tolerated dose was not reached. Forty-three patients (86.0%) had at least one treatment-related adverse event (TRAE), and three patients (6.0%) had ≥ G3 TRAEs. The most common TRAEs (any grade/grade ≥3) were AST increased (18%/0%), proteinuria (14%/2%), anemia (14%/2%), and ALT increased (12%/0%). No significant cardiotoxicity or bleeding was observed. GFH018 PK was linear and dose-independent, with a mean half-life of 2.25-8.60 h from 5 - 85 mg. Nine patients (18.0%) achieved stable disease, and one patient with thymic carcinoma achieved tumor shrinkage, with the maximum target lesion decreased by 18.4%. Serum TGF-β1 levels were not associated with clinical responses. The comprehensive recommended dose for Phase II was defined as 85 mg BID 14d-on/14d-off. CONCLUSIONS GFH018 monotherapy presented a favorable safety profile without cardiac toxicity or bleeding. Modest efficacy warrants further studies, including combination strategies. TRIAL REGISTRATION ClinicalTrial. gov ( https://www. CLINICALTRIALS gov/ ), NCT05051241. Registered on 2021-09-02.
Collapse
Affiliation(s)
- Ye Guo
- Department of Medical Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zishu Wang
- The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Huan Zhou
- The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Weidong Han
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yanhong Deng
- Sixth Affiliated Hospital of Sun Yat-Sen University, Guangdong, China
| | - Qun Li
- Department of Medical Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Junli Xue
- Department of Medical Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiaoxiao Ge
- Department of Medical Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shuang Wang
- Clinical Department, GenFleet Therapeutics Inc, Shanghai, China
| | - Jing Wang
- Translational Science, GenFleet Therapeutics Inc, Shanghai, China
| | - Yue Zhang
- Clinical Department, GenFleet Therapeutics Inc, Shanghai, China
| | - Congqiao Zhao
- Clinical Department, GenFleet Therapeutics Inc, Shanghai, China
| | - Huaqiang Zhu
- Clinical Department, GenFleet Therapeutics Inc, Shanghai, China
| | - Yu Wang
- Clinical Department, GenFleet Therapeutics Inc, Shanghai, China
| | - Haige Shen
- Clinical Department, GenFleet Therapeutics Inc, Shanghai, China
| | - Dong Liu
- Translational Science, GenFleet Therapeutics Inc, Shanghai, China
| | - Jin Li
- Department of Medical Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
8
|
Jiao YC, Wang YX, Liu WZ, Xu JW, Zhao YY, Yan CZ, Liu FC. Advances in the differentiation of pluripotent stem cells into vascular cells. World J Stem Cells 2024; 16:137-150. [PMID: 38455095 PMCID: PMC10915963 DOI: 10.4252/wjsc.v16.i2.137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/20/2023] [Accepted: 01/16/2024] [Indexed: 02/26/2024] Open
Abstract
Blood vessels constitute a closed pipe system distributed throughout the body, transporting blood from the heart to other organs and delivering metabolic waste products back to the lungs and kidneys. Changes in blood vessels are related to many disorders like stroke, myocardial infarction, aneurysm, and diabetes, which are important causes of death worldwide. Translational research for new approaches to disease modeling and effective treatment is needed due to the huge socio-economic burden on healthcare systems. Although mice or rats have been widely used, applying data from animal studies to human-specific vascular physiology and pathology is difficult. The rise of induced pluripotent stem cells (iPSCs) provides a reliable in vitro resource for disease modeling, regenerative medicine, and drug discovery because they carry all human genetic information and have the ability to directionally differentiate into any type of human cells. This review summarizes the latest progress from the establishment of iPSCs, the strategies for differentiating iPSCs into vascular cells, and the in vivo transplantation of these vascular derivatives. It also introduces the application of these technologies in disease modeling, drug screening, and regenerative medicine. Additionally, the application of high-tech tools, such as omics analysis and high-throughput sequencing, in this field is reviewed.
Collapse
Affiliation(s)
- Yi-Chang Jiao
- Department of Neurology, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
- Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Ying-Xin Wang
- Department of Neurology, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
- Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Wen-Zhu Liu
- Department of Neurology, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
- Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Jing-Wen Xu
- Department of Neurology, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
- Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Yu-Ying Zhao
- Department of Neurology, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
- Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Chuan-Zhu Yan
- Department of Neurology, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
- Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
- Mitochondrial Medicine Laboratory, Qilu Hospital (Qingdao) of Shandong University, Qingdao 266103, Shandong Province, China
- Brain Science Research Institute, Shandong University, Jinan 250012, Shandong Province, China
| | - Fu-Chen Liu
- Department of Neurology, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
- Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
- Brain Science Research Institute, Shandong University, Jinan 250012, Shandong Province, China.
| |
Collapse
|
9
|
Zhang L, Li Y, Yang W, Lin L, Li J, Liu D, Li C, Wu J, Li Y. Protocatechuic aldehyde increases pericyte coverage and mitigates pericyte damage to enhance the atherosclerotic plaque stability. Biomed Pharmacother 2023; 168:115742. [PMID: 37871558 DOI: 10.1016/j.biopha.2023.115742] [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: 08/14/2023] [Revised: 10/15/2023] [Accepted: 10/16/2023] [Indexed: 10/25/2023] Open
Abstract
Pericyte dysfunction and loss contribute substantially to the destabilization and rupture of atherosclerotic plaques. Protocatechuic aldehyde (PCAD), a natural polyphenol, exerts anti-atherosclerotic effects. However, the effects and mechanisms of this polyphenol on pericyte recruitment, coverage, and pericyte function remain unknown. We here treated apolipoprotein E-deficient mice having high-fat diet-induced atherosclerosis with PCAD. PCAD achieved therapeutic effects similar to rosuvastatin in lowering lipid levels and thus preventing atherosclerosis progression. With PCAD administration, plaque phenotype exhibited higher stability with markedly reduced lesion vulnerability, which is characterized by reduced lipid content and macrophage accumulation, and a consequent increase in collagen deposition. PCAD therapy increased pericyte coverage in the plaques, reduced VEGF-A production, and inhibited intraplaque neovascularization. PCAD promoted pericyte proliferation, adhesion, and migration to mitigate ox-LDL-induced pericyte dysfunction, which thus maintained the capillary network structure and stability. Furthermore, TGFBR1 silencing partially reversed the protective effect exerted by PCAD on human microvascular pericytes. PCAD increased pericyte coverage and impeded ox-LDL-induced damages through TGF-β1/TGFBR1/Smad2/3 signaling. All these novel findings indicated that PCAD increases pericyte coverage and alleviates pericyte damage to improve the stability of atherosclerotic plaques, which is accomplished by regulating TGF-β1/TGFBR1/Smad2/3 signaling in pericytes.
Collapse
Affiliation(s)
- Lei Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yuan Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Wenqing Yang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Lin Lin
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Jie Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Dekun Liu
- Shool of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Chao Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Jibiao Wu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Yunlun Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Department of Cardiovascular, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Shandong Provincial Engineering Laboratory of Traditional Chinese Medicine Precision Therapy for Cardiovascular Diseases, Jinan 250355, China.
| |
Collapse
|
10
|
Li K, Zeng X, Liu P, Zeng X, Lv J, Qiu S, Zhang P. The Role of Inflammation-Associated Factors in Head and Neck Squamous Cell Carcinoma. J Inflamm Res 2023; 16:4301-4315. [PMID: 37791117 PMCID: PMC10544098 DOI: 10.2147/jir.s428358] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/16/2023] [Indexed: 10/05/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC), which originates in the head or neck tissues, is characterized by high rates of recurrence and metastasis. Inflammation is important in HNSCC prognosis. Inflammatory cells and their secreted factors contribute to the various stages of HNSCC development through multiple mechanisms. In this review, the mechanisms through which inflammatory factors, signaling pathways, and cells contribute to the initiation and progression of HNSCC have been discussed in detail. Furthermore, the diagnostic and therapeutic potential of targeting inflammation in HNSCC has been discussed to gain new insights into improving patient prognosis.
Collapse
Affiliation(s)
- Kang Li
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zhuhai, Guangdong, People’s Republic of China
- Department of Otorhinolaryngology, Longgang Otorhinolaryngology Hospital & Shenzhen Key Laboratory of Otorhinolaryngology, Shenzhen Institute of Otorhinolaryngology, Shenzhen, Guangdong, People’s Republic of China
| | - Xianhai Zeng
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zhuhai, Guangdong, People’s Republic of China
- Department of Otorhinolaryngology, Longgang Otorhinolaryngology Hospital & Shenzhen Key Laboratory of Otorhinolaryngology, Shenzhen Institute of Otorhinolaryngology, Shenzhen, Guangdong, People’s Republic of China
| | - Peng Liu
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zhuhai, Guangdong, People’s Republic of China
- Department of Otorhinolaryngology, Longgang Otorhinolaryngology Hospital & Shenzhen Key Laboratory of Otorhinolaryngology, Shenzhen Institute of Otorhinolaryngology, Shenzhen, Guangdong, People’s Republic of China
| | - Xiaoxia Zeng
- Department of Otorhinolaryngology, Longgang Otorhinolaryngology Hospital & Shenzhen Key Laboratory of Otorhinolaryngology, Shenzhen Institute of Otorhinolaryngology, Shenzhen, Guangdong, People’s Republic of China
| | - Jie Lv
- School of Computer Science and Engineering, Yulin Normal University, Yulin, Guangxi, People’s Republic of China
| | - Shuqi Qiu
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zhuhai, Guangdong, People’s Republic of China
- Department of Otorhinolaryngology, Longgang Otorhinolaryngology Hospital & Shenzhen Key Laboratory of Otorhinolaryngology, Shenzhen Institute of Otorhinolaryngology, Shenzhen, Guangdong, People’s Republic of China
| | - Peng Zhang
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zhuhai, Guangdong, People’s Republic of China
- Department of Otorhinolaryngology, Longgang Otorhinolaryngology Hospital & Shenzhen Key Laboratory of Otorhinolaryngology, Shenzhen Institute of Otorhinolaryngology, Shenzhen, Guangdong, People’s Republic of China
| |
Collapse
|
11
|
Wang J, Song Y, Xie W, Zhao J, Wang Y, Yu W. Therapeutic angiogenesis based on injectable hydrogel for protein delivery in ischemic heart disease. iScience 2023; 26:106577. [PMID: 37192972 PMCID: PMC10182303 DOI: 10.1016/j.isci.2023.106577] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2023] Open
Abstract
Ischemic heart disease (IHD) remains the leading cause of death and disability worldwide and leads to myocardial necrosis and negative myocardial remodeling, ultimately leading to heart failure. Current treatments include drug therapy, interventional therapy, and surgery. However, some patients with severe diffuse coronary artery disease, complex coronary artery anatomy, and other reasons are unsuitable for these treatments. Therapeutic angiogenesis stimulates the growth of the original blood vessels by using exogenous growth factors to generate more new blood vessels, which provides a new treatment for IHD. However, direct injection of these growth factors can cause a short half-life and serious side effects owing to systemic spread. Therefore, to overcome this problem, hydrogels have been developed for temporally and spatially controlled delivery of single or multiple growth factors to mimic the process of angiogenesis in vivo. This paper reviews the mechanism of angiogenesis, some important bioactive molecules, and natural and synthetic hydrogels currently being applied for bioactive molecule delivery to treat IHD. Furthermore, the current challenges of therapeutic angiogenesis in IHD and its potential solutions are discussed to facilitate real translation into clinical applications in the future.
Collapse
Affiliation(s)
- Junke Wang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 26000, China
- Qingdao Medical College, Qingdao University, Qingdao, Shandong 266071, China
| | - Yancheng Song
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 26000, China
| | - Wenjie Xie
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Shandong, Qingdao, Shandong 26000, China
| | - Jiang Zhao
- Department of Urology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Ying Wang
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong 26000, China
- Corresponding author
| | - Wenzhou Yu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 26003, China
- Corresponding author
| |
Collapse
|
12
|
Díaz-García E, García-Sánchez A, Sánz-Rubio D, Alfaro E, López-Fernández C, Casitas R, Mañas Baena E, Cano-Pumarega I, Cubero P, Marin-Oto M, López-Collazo E, Marin JM, García-Río F, Cubillos-Zapata C. SMAD4 Expression in Monocytes as a Potential Biomarker for Atherosclerosis Risk in Patients with Obstructive Sleep Apnea. Int J Mol Sci 2023; 24:ijms24097900. [PMID: 37175608 PMCID: PMC10178665 DOI: 10.3390/ijms24097900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/14/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
Obstructive sleep apnea (OSA) patients are at special risk of suffering atherosclerosis, leading to major cardiovascular diseases. Notably, the transforming growth factor (TGF-β) plays a crucial role in the development and progression of atherosclerosis. In this context, the central regulator of TGF-β pathway, SMAD4 (small mother against decapentaplegic homolog 4), has been previously reported to be augmented in OSA patients, which levels were even higher in patients with concomitant cardiometabolic diseases. Here, we analyzed soluble and intracellular SMAD4 levels in plasma and monocytes from OSA patients and non-apneic subjects, with or without early subclinical atherosclerosis (eSA). In addition, we used in vitro and ex vivo models to explore the mechanisms underlying SMAD4 upregulation and release. Our study confirmed elevated sSMAD4 levels in OSA patients and identified that its levels were even higher in those OSA patients with eSA. Moreover, we demonstrated that SMAD4 is overexpressed in OSA monocytes and that intermittent hypoxia contributes to SMAD4 upregulation and release in a process mediated by NLRP3. In conclusion, this study highlights the potential role of sSMAD4 as a biomarker for atherosclerosis risk in OSA patients and provides new insights into the mechanisms underlying its upregulation and release to the extracellular space.
Collapse
Affiliation(s)
- Elena Díaz-García
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), 28029 Madrid, Spain
- Respiratory Diseases Group, Respiratory Diseases Department, La Paz University Hospital, IdiPAZ, 28046 Madrid, Spain
| | - Aldara García-Sánchez
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), 28029 Madrid, Spain
- Servicio de Neumología, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - David Sánz-Rubio
- Precision Medicine in Respiratory Diseases Group, Miguel Servet University Hospital-IIS Aragon, 50009 Zaragoza, Spain
| | - Enrique Alfaro
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), 28029 Madrid, Spain
- Respiratory Diseases Group, Respiratory Diseases Department, La Paz University Hospital, IdiPAZ, 28046 Madrid, Spain
| | - Cristina López-Fernández
- Respiratory Diseases Group, Respiratory Diseases Department, La Paz University Hospital, IdiPAZ, 28046 Madrid, Spain
| | - Raquel Casitas
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), 28029 Madrid, Spain
- Respiratory Diseases Group, Respiratory Diseases Department, La Paz University Hospital, IdiPAZ, 28046 Madrid, Spain
| | - Eva Mañas Baena
- Servicio de Neumología, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Irene Cano-Pumarega
- Servicio de Neumología, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Pablo Cubero
- Precision Medicine in Respiratory Diseases Group, Miguel Servet University Hospital-IIS Aragon, 50009 Zaragoza, Spain
| | - Marta Marin-Oto
- Precision Medicine in Respiratory Diseases Group, Miguel Servet University Hospital-IIS Aragon, 50009 Zaragoza, Spain
| | - Eduardo López-Collazo
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), 28029 Madrid, Spain
- The Innate Immune Response Group, La Paz University Hospital, IdiPAZ, 28046 Madrid, Spain
| | - José María Marin
- Precision Medicine in Respiratory Diseases Group, Miguel Servet University Hospital-IIS Aragon, 50009 Zaragoza, Spain
- Department of Medicine, University of Zaragoza School of Medicine, 50009 Zaragoza, Spain
| | - Francisco García-Río
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), 28029 Madrid, Spain
- Respiratory Diseases Group, Respiratory Diseases Department, La Paz University Hospital, IdiPAZ, 28046 Madrid, Spain
- Faculty of Medicine, Autonomous University of Madrid, 28029 Madrid, Spain
| | - Carolina Cubillos-Zapata
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), 28029 Madrid, Spain
- Respiratory Diseases Group, Respiratory Diseases Department, La Paz University Hospital, IdiPAZ, 28046 Madrid, Spain
| |
Collapse
|
13
|
Vaklavas C, Stringer-Reasor EM, Elkhanany AM, Ryan KJ, Li Y, Theuer CP, Acosta EP, Wei S, Yang ES, Grizzle WE, Forero-Torres A. A phase I/II study of preoperative letrozole, everolimus, and carotuximab in stage 2 and 3 hormone receptor-positive and Her2-negative breast cancer. Breast Cancer Res Treat 2023; 198:217-229. [PMID: 36735117 PMCID: PMC10020303 DOI: 10.1007/s10549-023-06864-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 01/16/2023] [Indexed: 02/04/2023]
Abstract
PURPOSE In nonmetastatic hormone receptor-positive and Her2-negative breast cancer, preoperative endocrine therapies can yield outcomes similar with chemotherapy. We evaluated the tolerability and preliminary antitumor activity of preoperative letrozole, everolimus, and carotuximab, a monoclonal antibody targeting endoglin, in nonmetastatic breast cancer. METHODS Eligible patients had newly diagnosed, stage 2 or 3, hormone receptor-positive and Her2/neu-negative breast cancer. Patients received escalating doses of everolimus; the dose of letrozole and carotuximab were fixed at 2.5 mg PO daily and 15 mg/kg intravenously every 2 weeks, respectively. The primary objective was to determine the safety and tolerability of the combination. Secondary objectives included pharmacokinetic and pharmacodynamic studies and assessments of antitumor activity. RESULTS Fifteen patients enrolled. The recommended phase 2 dose of everolimus in combination with letrozole and carotuximab was 10 mg PO daily. The most frequent adverse events were headache (67%), fatigue (47%), facial flushing and swelling (47%), gingival hemorrhage (40%), epistaxis (33%), nausea and vomiting (27%). Headache constituted a dose-limiting toxicity. At least two signs of mucocutaneous telangiectasia developed in 92% of patients. Carotuximab accumulated in the extravascular space and accelerated the biodistribution and clearance of everolimus. All patients had residual disease. Gene expression analyses were consistent with downregulation of genes involved in proliferation and DNA repair. Among 6 patients with luminal B breast cancer, 5 converted to luminal A after one cycle of therapy. CONCLUSION Letrozole, everolimus, and carotuximab were tolerated in combination at their single-agent doses. Pharmacokinetic studies revealed an interaction between everolimus and carotuximab. TRIAL REGISTRATION This trial is registered with ClinicalTrials.gov (Identifier: NCT02520063), first posted on August 11, 2015, and is active, not recruiting.
Collapse
Affiliation(s)
- Christos Vaklavas
- Huntsman Cancer Institute of the University of Utah, 2000 Circle of Hope, RS2509, Salt Lake, UT, 84112, USA.
- University of Alabama at Birmingham, Birmingham, AL, USA.
| | | | | | - Kevin J Ryan
- University of Alabama at Birmingham, Birmingham, AL, USA
| | - Yufeng Li
- University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | - Shi Wei
- University of Alabama at Birmingham, Birmingham, AL, USA
| | - Eddy S Yang
- University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | |
Collapse
|
14
|
Extracellular Vesicles Secreted by TGF-β1-Treated Mesenchymal Stem Cells Promote Fracture Healing by SCD1-Regulated Transference of LRP5. Stem Cells Int 2023; 2023:4980871. [PMID: 36970598 PMCID: PMC10033213 DOI: 10.1155/2023/4980871] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 08/26/2022] [Accepted: 02/03/2023] [Indexed: 03/17/2023] Open
Abstract
Bone fracture repair is a multiphased regenerative process requiring paracrine intervention throughout the healing process. Mesenchymal stem cells (MSCs) play a crucial role in cell-to-cell communication and the regeneration of tissue, but their transplantation is difficult to regulate. The paracrine processes that occur in MSC-derived extracellular vesicles (MSC-EVs) have been exploited for this study. The primary goal was to determine whether EVs secreted by TGF-β1-stimulated MSCs (MSCTGF-β1-EVs) exhibit greater effects on bone fracture healing than EVs secreted by PBS-treated MSCs (MSCPBS-EVs). Our research was conducted using an in vivo bone fracture model and in vitro experiments, which included assays to measure cell proliferation, migration, and angiogenesis, as well as in vivo and in vitro gain/loss of function studies. In this study, we were able to confirm that SCD1 expression and MSC-EVs can be induced by TGF-β1. After MSCTGF-β1-EVs are transplanted in mice, bone fracture repair is accelerated. MSCTGF-β1-EV administration stimulates human umbilical vein endothelial cell (HUVEC) angiogenesis, proliferation, and migration in vitro. Furthermore, we were able to demonstrate that SCD1 plays a functional role in the process of MSCTGF-β1-EV-mediated bone fracture healing and HUVEC angiogenesis, proliferation, and migration. Additionally, using a luciferase reporter assay and chromatin immunoprecipitation studies, we discovered that SREBP-1 targets the promoter of the SCD1 gene specifically. We also discovered that the EV-SCD1 protein could stimulate proliferation, angiogenesis, and migration in HUVECs through interactions with LRP5. Our findings provide evidence of a mechanism whereby MSCTGF-β1-EVs enhance bone fracture repair by regulating the expression of SCD1. The use of TGF-β1 preconditioning has the potential to maximize the therapeutic effects of MSC-EVs in the treatment of bone fractures.
Collapse
|
15
|
Shineh G, Patel K, Mobaraki M, Tayebi L. Functional Approaches in Promoting Vascularization and Angiogenesis in Bone Critical-Sized Defects via Delivery of Cells, Growth Factors, Drugs, and Particles. J Funct Biomater 2023; 14:99. [PMID: 36826899 PMCID: PMC9960138 DOI: 10.3390/jfb14020099] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023] Open
Abstract
Critical-sized bone defects, or CSDs, are defined as bone defects that cannot be regenerated by themselves and require surgical intervention via employing specific biomaterials and a certain regenerative strategy. Although a variety of approaches can be used to treat CSDs, poor angiogenesis and vascularization remain an obstacle in these methods. The complex biological healing of bone defects depends directly on the function of blood flow to provide sufficient oxygen and nutrients and the removal of waste products from the defect site. The absence of vascularization can lead to non-union and delayed-union defect development. To overcome this challenge, angiogenic agents can be delivered to the site of injury to stimulate vessel formation. This review begins by introducing the treatment methods for CSDs. The importance of vascularization in CSDs is subsequently highlighted. Delivering angiogenesis agents, including relevant growth factors, cells, drugs, particles, cell secretion substances, their combination, and co-delivery to CSDs are fully explored. Moreover, the effects of such agents on new bone formation, followed by vessel formation in defect areas, are evaluated.
Collapse
Affiliation(s)
- Ghazal Shineh
- School of Biomedical Engineering, University of Sydney, Sydney, NSW 2006, Australia
| | - Kishan Patel
- School of Dentistry, Marquette University, Milwaukee, WI 53207, USA
| | - Mohammadmahdi Mobaraki
- Biomaterial Group, Faculty of Biomedical Engineering, Amirkabir University of Technology, Tehran 15916-34311, Iran
| | - Lobat Tayebi
- School of Dentistry, Marquette University, Milwaukee, WI 53207, USA
| |
Collapse
|
16
|
Feng J, Tang D, Wang J, Zhou Q, Peng J, Lou H, Sun Y, Cai Y, Chen H, Yang J, Liu P, Wang L, Zou J. SHR-1701, a Bifunctional Fusion Protein Targeting PD-L1 and TGFβ, for Recurrent or Metastatic Cervical Cancer: A Clinical Expansion Cohort of a Phase I Study. Clin Cancer Res 2022; 28:5297-5305. [PMID: 35653122 DOI: 10.1158/1078-0432.ccr-22-0346] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/20/2022] [Accepted: 05/27/2022] [Indexed: 01/24/2023]
Abstract
PURPOSE Patients with recurrent or metastatic cervical cancer have limited treatment options after platinum-containing treatment. We initiated a phase I study to assess SHR-1701, a novel bifunctional fusion protein composed of a mAb against programmed death ligand 1 (PD-L1) fused with the extracellular domain of TGFβ receptor II, in solid tumors (NCT03774979). Here, results from the cervical cancer cohort are presented. PATIENTS AND METHODS Patients with recurrent or metastatic cervical cancer who progressed during or after platinum-based therapy were enrolled to receive SHR-1701 at 30 mg/kg every 3 weeks. Primary endpoint was objective response rate (ORR) per RECIST v1.1. RESULTS In total, 32 patients were recruited. ORR was 15.6% [95% confidence interval (CI), 5.3-32.8], and disease control rate was 50.0% (95% CI, 31.9-68.1). Responses were still ongoing in 80.0% of the responders; 6-month duration of response rate was 80.0% (95% CI, 20.4-96.9). Median progression-free survival (PFS) was 2.7 months (95% CI, 1.4-4.1). Of note, as assessed by immune-modified RECIST, median PFS was 4.1 months (95% CI, 1.6-4.3). Overall survival rate at 12 months was 54.6% (95% CI, 31.8-72.7). Treatment-related adverse events of grade 3 or 4 were reported in 11 (34.4%) patients. No treatment-related deaths occurred. No difference in ORR was found between patients with PD-L1 combined positive score ≥1 or <1; patients with high phosphorylated SMAD2 level in immune cells or tumor cells had numerically higher ORR. CONCLUSIONS SHR-1701 exhibits encouraging antitumor activity and controllable safety in patients with recurrent or metastatic cervical cancer after platinum-based regimens, and therefore might provide another treatment option for this population. See related commentary by Miller and Friedman, p. 5238.
Collapse
Affiliation(s)
- Jifeng Feng
- Department of Oncology, Jiangsu Cancer Hospital, Nanjing, P.R. China
| | - Dihong Tang
- Gynecologic Oncology, Hunan Cancer Hospital, Changsha, P.R. China
| | - Jing Wang
- Gynecologic Oncology, Hunan Cancer Hospital, Changsha, P.R. China
| | - Qi Zhou
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital, Chongqing, P.R. China
| | - Jin Peng
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, P.R. China
| | - Hanmei Lou
- Gynecological Surgery, Zhejiang Cancer Hospital, Hangzhou, P.R. China
| | - Yuping Sun
- Medical Oncology, Jinan Central Hospital, Jinan, P.R. China
| | - Yunlang Cai
- Department of Gynecology and Obstetrics, Zhongda Hospital Southeast University, Nanjing, P.R. China
| | - Hongmin Chen
- Gynecology, Henan Cancer Hospital, Zhengzhou, P.R. China
| | - Junqin Yang
- Clinical Research and Development, Jiangsu Hengrui Pharmaceuticals Co., Ltd, Shanghai, P.R. China
| | - Pan Liu
- Clinical Research and Development, Jiangsu Hengrui Pharmaceuticals Co., Ltd, Shanghai, P.R. China
| | - Linna Wang
- Clinical Research and Development, Jiangsu Hengrui Pharmaceuticals Co., Ltd, Shanghai, P.R. China
| | - Jianjun Zou
- Clinical Research and Development, Jiangsu Hengrui Pharmaceuticals Co., Ltd, Shanghai, P.R. China
| |
Collapse
|
17
|
Wang Y, Wang G, Liu H. Tenascin-C: A Key Regulator in Angiogenesis during Wound Healing. Biomolecules 2022; 12:1689. [PMID: 36421704 PMCID: PMC9687801 DOI: 10.3390/biom12111689] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 08/27/2023] Open
Abstract
(1) Background: Injury repair is a complex physiological process in which multiple cells and molecules are involved. Tenascin-C (TNC), an extracellular matrix (ECM) glycoprotein, is essential for angiogenesis during wound healing. This study aims to provide a comprehensive review of the dynamic changes and functions of TNC throughout tissue regeneration and to present an up-to-date synthesis of the body of knowledge pointing to multiple mechanisms of TNC at different restoration stages. (2) Methods: A review of the PubMed database was performed to include all studies describing the pathological processes of damage restoration and the role, structure, expression, and function of TNC in post-injury treatment; (3) Results: In this review, we first introduced the construction and expression signature of TNC. Then, the role of TNC during the process of damage restoration was introduced. We highlight the temporal heterogeneity of TNC levels at different restoration stages. Furthermore, we are surprised to find that post-injury angiogenesis is dynamically consistent with changes in TNC. Finally, we discuss the strategies for TNC in post-injury treatment. (4) Conclusions: The dynamic expression of TNC has a significant impact on angiogenesis and healing wounds and counters many negative aspects of poorly healing wounds, such as excessive inflammation, ischemia, scarring, and wound infection.
Collapse
Affiliation(s)
- Yucai Wang
- Department of Orthopaedic Surgery, Tangdu Hospital, AirForce Medical University, Xi’an 710000, China
| | - Guangfu Wang
- Vasculocardiology Department, The Fourth People’s Hospital of Jinan, Jinan 250000, China
| | - Hao Liu
- Division of Vascular and Interventional Radiology, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| |
Collapse
|
18
|
Liu D, Zhou J, Wang Y, Li M, Jiang H, Liu Y, Yin X, Ge M, Xiang X, Ying J, Huang J, Zhang YQ, Cheng Y, Huang Z, Yuan X, Han W, Yan D, Wang X, Liu P, Wang L, Zhang X, Luo S, Liu T, Shen L. Bifunctional anti-PD-L1/TGF-βRII agent SHR-1701 in advanced solid tumors: a dose-escalation, dose-expansion, and clinical-expansion phase 1 trial. BMC Med 2022; 20:408. [PMID: 36280870 PMCID: PMC9594927 DOI: 10.1186/s12916-022-02605-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 10/12/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Dual inhibition of PD-1/PD-L1 and TGF-β pathways is a rational therapeutic strategy for malignancies. SHR-1701 is a new bifunctional fusion protein composed of a monoclonal antibody against PD-L1 fused with the extracellular domain of TGF-β receptor II. This first-in-human trial aimed to assess SHR-1701 in pretreated advanced solid tumors and find the population who could benefit from SHR-1701. METHODS This was a dose-escalation, dose-expansion, and clinical-expansion phase 1 study. Dose escalation was initiated by accelerated titration (1 mg/kg q3w; intravenous infusion) and then switched to a 3+3 scheme (3, 10, 20, and 30 mg/kg q3w and 30 mg/kg q2w), followed by dose expansion at 10, 20, and 30 mg/kg q3w and 30 mg/kg q2w. The primary endpoints of the dose-escalation and dose-expansion parts were the maximum tolerated dose and recommended phase 2 dose. In the clinical-expansion part, selected tumors were enrolled to receive SHR-1701 at the recommended dose, with a primary endpoint of confirmed objective response rate (ORR). RESULTS In total, 171 patients were enrolled (dose-escalation: n=17; dose-expansion, n=33; clinical-expansion, n=121). In the dose-escalation part, no dose-limiting toxicity was observed, and the maximum tolerated dose was not reached. SHR-1701 showed a linear dose-exposure relationship and the highest ORR at 30 mg/kg every 3 weeks, without obviously aggravated toxicities across doses in the dose-escalation and dose-expansion parts. Combined, 30 mg/kg every 3 weeks was determined as the recommended phase 2 dose. In the clinical-expansion part, SHR-1701 showed the most favorable efficacy in the gastric cancer cohort, with an ORR of 20.0% (7/35; 95% CI, 8.4-36.9) and a 12-month overall survival rate of 54.5% (95% CI, 29.5-73.9). Grade ≥3 treatment-related adverse events occurred in 37 of 171 patients (22%), mainly including increased gamma-glutamyltransferase (4%), increased aspartate aminotransferase (3%), anemia (3%), hyponatremia (3%), and rash (2%). Generally, patients with PD-L1 CPS ≥1 or pSMAD2 histochemical score ≥235 had numerically higher ORR. CONCLUSIONS SHR-1701 showed an acceptable safety profile and encouraging antitumor activity in pretreated advanced solid tumors, especially in gastric cancer, establishing the foundation for further exploration. TRIAL REGISTRATION ClinicalTrials.gov , NCT03710265.
Collapse
Affiliation(s)
- Dan Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Early Drug Development Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jun Zhou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Fu-Cheng Road 52, Hai-Dian District, Beijing, 100142, China
| | - Yongsheng Wang
- Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Mingjun Li
- Oncology Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Haiping Jiang
- Department of Medical Oncology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yunpeng Liu
- Medical Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Xianli Yin
- Department of Gastroenterology, Hunan Cancer Hospital, Changsha, China
| | - Minghua Ge
- Head and Neck Surgery, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Xiaojun Xiang
- Oncology Department, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jieer Ying
- Hepatobiliary Pancreatic Gastroenterology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Jian Huang
- Urinary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yan-Qiao Zhang
- Ward 2, Department of Gastroenterology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ying Cheng
- Oncology Department, Jilin Cancer Hospital, Changchun, China
| | - Zhigang Huang
- Otolaryngology Head and Neck Surgery Center, Beijing Tongren Hospital, Beijing, China
| | - Xianglin Yuan
- Oncology Department, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weiqing Han
- Urology Surgery, Hunan Cancer Hospital (The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University), Changsha, China
| | - Dong Yan
- Oncology Department, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Xinshuai Wang
- Oncology Department, Henan Key Laboratory of Cancer Epigenetics, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China
| | - Pan Liu
- Clinical Research & Development, Jiangsu Hengrui Pharmaceuticals Co., Ltd, Shanghai, China
| | - Linna Wang
- Clinical Research & Development, Jiangsu Hengrui Pharmaceuticals Co., Ltd, Shanghai, China
| | - Xiaojing Zhang
- Clinical Research & Development, Jiangsu Hengrui Pharmaceuticals Co., Ltd, Shanghai, China
| | - Suxia Luo
- Department of Gastroenterology, Henan Tumor Hospital, Dongming Road 127, Jinshui District, Zhengzhou, 450003, China.
| | - Tianshu Liu
- Medical Oncology, Zhongshan Hospital Fudan University, Fenglin Road 180, Xuhui District, Shanghai, 200032, China.
| | - Lin Shen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Fu-Cheng Road 52, Hai-Dian District, Beijing, 100142, China.
| |
Collapse
|
19
|
Făgărășan A, Săsăran MO. The Predictive Role of Plasma Biomarkers in the Evolution of Aortopathies Associated with Congenital Heart Malformations. Int J Mol Sci 2022; 23:ijms23094993. [PMID: 35563383 PMCID: PMC9102091 DOI: 10.3390/ijms23094993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 02/06/2023] Open
Abstract
Dilatation of the aorta is a constantly evolving condition that can lead to the ultimate life-threatening event, acute aortic dissection. Recent research has tried to identify quantifiable biomarkers, with both diagnostic and prognostic roles in different aortopathies. Most studies have focused on the bicuspid aortic valve, the most frequent congenital heart disease (CHD), and majorly evolved around matrix metalloproteinases (MMPs). Other candidate biomarkers, such as asymmetric dimethylarginine, soluble receptor for advanced glycation end-products or transforming growth factor beta have also gained a lot of attention recently. Most of the aortic anomalies and dilatation-related studies have reported expression variation of tissular biomarkers. The ultimate goal remains, though, the identification of biomarkers among the serum plasma, with the upregulation of circulating MMP-1, MMP-2, MMP-9, tissue inhibitor of metalloproteinase-1 (TIMP-1), asymmetric dimethylarginine (ADMA), soluble receptor for advanced glycation end-products (sRAGE) and transforming growth factor beta (TGF-β) being reported in association to several aortopathies and related complications in recent research. These molecules are apparently quantifiable from the early ages and have been linked to several CHDs and hereditary aortopathies. Pediatric data on the matter is still limited, and further studies are warranted to elucidate the role of plasmatic biomarkers in the long term follow-up of potentially evolving congenital aortopathies.
Collapse
Affiliation(s)
- Amalia Făgărășan
- Department of Pediatrics III, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540136 Târgu Mureș, Romania;
| | - Maria Oana Săsăran
- Department of Pediatrics III, Faculty of Medicine in English, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540136 Târgu Mureș, Romania
- Correspondence: ; Tel.: +40-720-332-503
| |
Collapse
|
20
|
Liu M, Yang J, Xu B, Zhang X. Tumor metastasis: Mechanistic insights and therapeutic interventions. MedComm (Beijing) 2021; 2:587-617. [PMID: 34977870 PMCID: PMC8706758 DOI: 10.1002/mco2.100] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/01/2021] [Accepted: 11/03/2021] [Indexed: 12/18/2022] Open
Abstract
Cancer metastasis is responsible for the vast majority of cancer-related deaths worldwide. In contrast to numerous discoveries that reveal the detailed mechanisms leading to the formation of the primary tumor, the biological underpinnings of the metastatic disease remain poorly understood. Cancer metastasis is a complex process in which cancer cells escape from the primary tumor, settle, and grow at other parts of the body. Epithelial-mesenchymal transition and anoikis resistance of tumor cells are the main forces to promote metastasis, and multiple components in the tumor microenvironment and their complicated crosstalk with cancer cells are closely involved in distant metastasis. In addition to the three cornerstones of tumor treatment, surgery, chemotherapy, and radiotherapy, novel treatment approaches including targeted therapy and immunotherapy have been established in patients with metastatic cancer. Although the cancer survival rate has been greatly improved over the years, it is still far from satisfactory. In this review, we provided an overview of the metastasis process, summarized the cellular and molecular mechanisms involved in the dissemination and distant metastasis of cancer cells, and reviewed the important advances in interventions for cancer metastasis.
Collapse
Affiliation(s)
- Mengmeng Liu
- Melanoma and Sarcoma Medical Oncology UnitState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Jing Yang
- Melanoma and Sarcoma Medical Oncology UnitState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Bushu Xu
- Melanoma and Sarcoma Medical Oncology UnitState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Xing Zhang
- Melanoma and Sarcoma Medical Oncology UnitState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| |
Collapse
|
21
|
Connective Tissue Disorders and Cardiovascular Complications: The Indomitable Role of Transforming Growth Factor-β Signaling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1348:161-184. [PMID: 34807419 DOI: 10.1007/978-3-030-80614-9_7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Marfan Syndrome (MFS) and Loeys-Dietz Syndrome (LDS) represent heritable connective tissue disorders that segregate with a similar pattern of cardiovascular defects (thoracic aortic aneurysm, mitral valve prolapse/regurgitation, and aortic dilatation with regurgitation). This pattern of cardiovascular defects appears to be expressed along a spectrum of severity in many heritable connective tissue disorders and raises suspicion of a relationship between the normal development of connective tissues and the cardiovascular system. With overwhelming evidence of the involvement of aberrant Transforming Growth Factor-beta (TGF-β) signaling in MFS and LDS, this signaling pathway may represent the common link in the relationship between connective tissue disorders and their associated cardiovascular complications. To further explore this hypothetical link, this chapter will review the TGF-β signaling pathway, the heritable connective tissue syndromes related to aberrant TGF-β signaling, and will discuss the pathogenic contribution of TGF-β to these syndromes with a primary focus on the cardiovascular system.
Collapse
|
22
|
The Dual Effect of the BMP9-ALK1 Pathway in Blood Vessels: An Opportunity for Cancer Therapy Improvement? Cancers (Basel) 2021; 13:cancers13215412. [PMID: 34771575 PMCID: PMC8582496 DOI: 10.3390/cancers13215412] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary The modulation of tumor blood vessels is a great opportunity for improving cancer therapies. Understanding the cellular and molecular players that regulate the biology of tumor blood vessels and tumor angiogenesis is necessary for the development of new anti-tumor strategies. Bone morphogenetic protein 9 (BMP9) is a circulating factor with multiple effects in vascular biology through its receptor activin receptor-like kinase 1 (ALK1). In this review, we give an overview of the possible benefits of modulating BMP9–ALK1 functions for cancer therapy improvement. Abstract The improvement of cancer therapy efficacy, the extension of patient survival and the reduction of adverse side effects are major challenges in cancer research. Targeting blood vessels has been considered a promising strategy in cancer therapy. Since the tumor vasculature is disorganized, leaky and triggers immunosuppression and tumor hypoxia, several strategies have been studied to modify tumor vasculature for cancer therapy improvement. Anti-angiogenesis was first described as a mechanism to prevent the formation of new blood vessels and prevent the oxygen supply to tumor cells, showing numerous limitations. Vascular normalization using low doses of anti-angiogenic drugs was purposed to overcome the limitations of anti-angiogenic therapies. Other strategies such as vascular promotion or the induction of high endothelial venules are being studied now to improve cancer therapy. Bone morphogenetic protein 9 (BMP9) exerts a dual effect through the activin receptor-like kinase 1 (ALK1) receptor in blood vessel maturation or activation phase of angiogenesis. Thus, it is an interesting pathway to target in combination with chemotherapies or immunotherapies. This review manuscript explores the effect of the BMP9–ALK1 pathway in tumor angiogenesis and the possible usefulness of targeting this pathway in anti-angiogenesis, vascular normalization or vascular promotion therapies.
Collapse
|
23
|
Briasoulis A, Ruiz Duque E, Mouselimis D, Tsarouchas A, Bakogiannis C, Alvarez P. The role of renin-angiotensin system in patients with left ventricular assist devices. J Renin Angiotensin Aldosterone Syst 2021; 21:1470320320966445. [PMID: 33084480 PMCID: PMC7871286 DOI: 10.1177/1470320320966445] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
End-stage heart failure is a condition in which the up-regulation of the systemic and local renin-angiotensin-aldosterone system (RAAS) leads to end-organ damage and is largely irreversible despite optimal medication. Left ventricular assist devices (LVADs) can downregulate RAAS activation by unloading the left ventricle and increasing the cardiac output translating into a better end-organ perfusion improving survival. However, the absence of pulsatility brought about by continuous-flow devices may variably trigger RAAS activation depending on left ventricular (LV) intrinsic contractility, the design and speed of the pump device. Moreover, the concept of myocardial recovery is being tested in clinical trials and in this setting LVAD support combined with intense RAAS inhibition can promote recovery and ensure maintenance of LV function after explantation. Blood pressure control on LVAD recipients is key to avoiding complications as gastrointestinal bleeding, pump thrombosis and stroke. Furthermore, emerging data highlight the role of RAAS antagonists as prevention of arteriovenous malformations that lead to gastrointestinal bleeds. Future studies should focus on the role of angiotensin receptor inhibitors in preventing myocardial fibrosis in patients with LVADs and examine in greater details the target blood pressure for these patients.
Collapse
Affiliation(s)
- Alexandros Briasoulis
- Division of Cardiovascular Diseases, Section of Heart Failure and Transplant, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Ernesto Ruiz Duque
- Division of Cardiovascular Diseases, Section of Heart Failure and Transplant, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Dimitrios Mouselimis
- 3rd Department of Cardiology Hippocration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Anastasios Tsarouchas
- 3rd Department of Cardiology Hippocration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Constantinos Bakogiannis
- Division of Cardiovascular Diseases, Section of Heart Failure and Transplant, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Paulino Alvarez
- Division of Cardiovascular Diseases, Section of Heart Failure and Transplant, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| |
Collapse
|
24
|
Babaei M, Sarhangi N, Shahrabi-Farahani M, Sharifi F, Alipor M, Aminimoghaddam S, Amoli MM. Investigation of TGF-β1 gene variant and expression in a group of Iranian women with endometriosis. Arch Gynecol Obstet 2021; 304:1527-1534. [PMID: 34480227 DOI: 10.1007/s00404-021-06195-4] [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: 11/29/2020] [Accepted: 08/17/2021] [Indexed: 11/24/2022]
Abstract
PURPOSE Endometriosis is defined as a common gynecologic and inflammatory disease. Transforming growth factor-beta 1 (TGF-β1) gene and its protein level might play a role in the pathogenesis of endometriosis. The present study aimed for the first time to assess the associations between endometriosis risk and - 509 C/T (rs1800469) variant of the TGF-β1 gene as well as TGF-β1 mRNA expression in eutopic endometrium tissue of patients with and without endometriosis among a group of Iranian women. METHODS Genotyping was carried out in 100 endometriosis patients (cases) with confirmed histological diagnosis of endometriosis and 197 non-endometriosis subjects (controls). The expression level of TGF-β1 mRNA was determined using Real-Time PCR assay in 15 eutopic endometrium tissue of women with endometriosis and 15 healthy controls. RESULTS There was a significant association for allele and genotype frequencies of rs1800469 variant and endometriosis. No significant difference for TGF-β1 expression was observed between eutopic endometrium of patients and healthy group. Also, evaluation of TGF-β1expression across the menstrual cycle showed the same level of TGF-β1 among case and control subjects. CONCLUSION Our investigations indicated enough evidence for the effect of TGF-β1 genetic variant on endometriosis risk in an Iranian population. Furthermore, we could not find any relations between TGF-β1 mRNA expression and susceptibility to endometriosis.
Collapse
Affiliation(s)
- Mahdokht Babaei
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.,Department Cell and Molecular Biology, Babol Branch, Islamic Azad University, Babol, Iran
| | - Negar Sarhangi
- Personalized Medicine Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, No.10- Jalal -e-Ale-Ahmad Street, Chamran Highway, 1411713119, Tehran, Iran.
| | - Maryam Shahrabi-Farahani
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Farshad Sharifi
- Elderly Health Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Majid Alipor
- Department Cell and Molecular Biology, Babol Branch, Islamic Azad University, Babol, Iran
| | - Soheila Aminimoghaddam
- Department of Gynecology and Oncology, Iran University of Medical Sciences, Shahid Hemmat Highway, 1449614535, Tehran, Iran.
| | - Mahsa M Amoli
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.,Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
25
|
Kant V, Jangir BL, Sharma M, Kumar V, Joshi VG. Topical application of quercetin improves wound repair and regeneration in diabetic rats. Immunopharmacol Immunotoxicol 2021; 43:536-553. [PMID: 34278923 DOI: 10.1080/08923973.2021.1950758] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Purpose: There is an urgent need of effective drug/formulation to speed up the healing process in diabetic wounds. In our earlier studies, quercetin has accelerated the healing of nondiabetic wounds. So, we investigated the wound-healing potentials of quercetin in diabetic rats.Materials and methods: A square-shaped cutaneous wound (≈400 mm2) was created on the back of nondiabetic and diabetic rats. They were divided into three groups, viz. healthy control (nondiabetic), diabetic control and diabetic-treated group. Ointment base was topically applied for 21 days in healthy and diabetic control groups. Quercetin (0.3%) ointment was similarly applied in third group. Effects of quercetin on repair and regenerations of diabetic wounds in terms of wound closure, inflammation, angiogenesis, fibroblast proliferation, collagen synthesis, epithelialization, axonal regeneration etc was studied.Results: Quercetin accelerated the wound closure and increased the expressions of IL-10, VEGF and TGF-β1 in granulation/healing tissue of diabetic wound. However, quercetin decreased the expression of TNF-α, IL-1β, and MMP-9. Histopathological evaluation revealed amelioration of persistence of inflammatory cells by quercetin in diabetic wounds. There was good quality of granulation tissue, marked fibroblast proliferation, well organized collagen deposition, early regeneration of epithelial layer etc. in the quercetin treated diabetic wounds in comparison to diabetic control group. Results of immunohistochemistry showed more angiogenesis, faster phenotypic switching of fibroblast to myofibroblasts and increased GAP-43 positive nerve fibers in quercetin-treated diabetic wounds.Conclusion: Quercetin ointment at 0.3% w/w concentration modulates cytokines, growth factors and protease, thereby improved repair and regenerations of cutaneous diabetic wounds in rats.
Collapse
Affiliation(s)
- Vinay Kant
- Department of Veterinary Pharmacology and Toxicology, Lala Lajpat Rai University of Veterinary & Animal Sciences (LUVAS), Hisar, Haryana, India
| | - Babu Lal Jangir
- Department of Veterinary Pathology, Lala Lajpat Rai University of Veterinary & Animal Sciences (LUVAS), Hisar, Haryana, India
| | - Maneesh Sharma
- Department of Veterinary Clinical Complex, College of Veterinary Sciences, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, Haryana, India
| | - Vinod Kumar
- Department of Veterinary Pharmacology and Toxicology, Lala Lajpat Rai University of Veterinary & Animal Sciences (LUVAS), Hisar, Haryana, India
| | - Vinay G Joshi
- Department of Animal Biotechnology, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, Haryana, India
| |
Collapse
|
26
|
Yuan K, Agarwal S, Chakraborty A, Condon DF, Patel H, Zhang S, Huang F, Mello SA, Kirk OI, Vasquez R, de Jesus Perez VA. Lung Pericytes in Pulmonary Vascular Physiology and Pathophysiology. Compr Physiol 2021; 11:2227-2247. [PMID: 34190345 PMCID: PMC10507675 DOI: 10.1002/cphy.c200027] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Pericytes are mesenchymal-derived mural cells localized within the basement membrane of pulmonary and systemic capillaries. Besides structural support, pericytes control vascular tone, produce extracellular matrix components, and cytokines responsible for promoting vascular homeostasis and angiogenesis. However, pericytes can also contribute to vascular pathology through the production of pro-inflammatory and pro-fibrotic cytokines, differentiation into myofibroblast-like cells, destruction of the extracellular matrix, and dissociation from the vessel wall. In the lung, pericytes are responsible for maintaining the integrity of the alveolar-capillary membrane and coordinating vascular repair in response to injury. Loss of pericyte communication with alveolar capillaries and a switch to a pro-inflammatory/pro-fibrotic phenotype are common features of lung disorders associated with vascular remodeling, inflammation, and fibrosis. In this article, we will address how to differentiate pericytes from other cells, discuss the molecular mechanisms that regulate the interactions of pericytes and endothelial cells in the pulmonary circulation, and the experimental tools currently used to study pericyte biology both in vivo and in vitro. We will also discuss evidence that links pericytes to the pathogenesis of clinically relevant lung disorders such as pulmonary hypertension, idiopathic lung fibrosis, sepsis, and SARS-COVID. Future studies dissecting the complex interactions of pericytes with other pulmonary cell populations will likely reveal critical insights into the origin of pulmonary diseases and offer opportunities to develop novel therapeutics to treat patients afflicted with these devastating disorders. © 2021 American Physiological Society. Compr Physiol 11:2227-2247, 2021.
Collapse
Affiliation(s)
- Ke Yuan
- Division of Respiratory Diseases Research, Department of Pediatrics, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Stuti Agarwal
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Ananya Chakraborty
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - David F. Condon
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Hiral Patel
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Serena Zhang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Flora Huang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Salvador A. Mello
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | | | - Rocio Vasquez
- University of Central Florida, Orlando, Florida, USA
| | - Vinicio A. de Jesus Perez
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| |
Collapse
|
27
|
Inhibiting Endothelial Cell Function in Normal and Tumor Angiogenesis Using BMP Type I Receptor Macrocyclic Kinase Inhibitors. Cancers (Basel) 2021; 13:cancers13122951. [PMID: 34204675 PMCID: PMC8231556 DOI: 10.3390/cancers13122951] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 06/07/2021] [Accepted: 06/09/2021] [Indexed: 01/05/2023] Open
Abstract
Angiogenesis, i.e., the formation of new blood vessels from pre-existing endothelial cell (EC)-lined vessels, is critical for tissue development and also contributes to neovascularization-related diseases, such as cancer. Vascular endothelial growth factor (VEGF) and bone morphogenetic proteins (BMPs) are among many secreted cytokines that regulate EC function. While several pharmacological anti-angiogenic agents have reached the clinic, further improvement is needed to increase clinical efficacy and to overcome acquired therapy resistance. More insights into the functional consequences of targeting specific pathways that modulate blood vessel formation may lead to new therapeutic approaches. Here, we synthesized and identified two macrocyclic small molecular compounds termed OD16 and OD29 that inhibit BMP type I receptor (BMPRI)-induced SMAD1/5 phosphorylation and downstream gene expression in ECs. Of note, OD16 and OD29 demonstrated higher specificity against BMPRI activin receptor-like kinase 1/2 (ALK1/2) than the commonly used small molecule BMPRI kinase inhibitor LDN-193189. OD29, but not OD16, also potently inhibited VEGF-induced extracellular regulated kinase MAP kinase phosphorylation in ECs. In vitro, OD16 and OD29 exerted strong inhibition of BMP9 and VEGF-induced ECs migration, invasion and cord formation. Using Tg (fli:EGFP) zebrafish embryos, we found that OD16 and OD29 potently antagonized dorsal longitudinal anastomotic vessel (DLAV), intra segmental vessel (ISV), and subintestinal vessel (SIV) formation during embryonic development. Moreover, the MDA-MB-231 breast cancer cell-induced tumor angiogenesis in zebrafish embryos was significantly decreased by OD16 and OD29. Both macrocyclic compounds might provide a steppingstone for the development of novel anti-angiogenesis therapeutic agents.
Collapse
|
28
|
Zhu H, Zhang Y, Zhong Y, Ye Y, Hu X, Gu L, Xiong X. Inflammation-Mediated Angiogenesis in Ischemic Stroke. Front Cell Neurosci 2021; 15:652647. [PMID: 33967696 PMCID: PMC8096981 DOI: 10.3389/fncel.2021.652647] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 02/16/2021] [Indexed: 12/12/2022] Open
Abstract
Stroke is the leading cause of disability and mortality in the world, but the pathogenesis of ischemic stroke (IS) is not completely clear and treatments are limited. Mounting evidence indicate that neovascularization is a critical defensive reaction to hypoxia that modulates the process of long-term neurologic recovery after IS. Angiogenesis is a complex process in which the original endothelial cells in blood vessels are differentiated, proliferated, migrated, and finally remolded into new blood vessels. Many immune cells and cytokines, as well as growth factors, are directly or indirectly involved in the regulation of angiogenesis. Inflammatory cells can affect endothelial cell proliferation, migration, and activation by secreting a variety of cytokines via various inflammation-relative signaling pathways and thus participate in the process of angiogenesis. However, the mechanism of inflammation-mediated angiogenesis has not been fully elucidated. Hence, this review aimed to discuss the mechanism of inflammation-mediated angiogenesis in IS and to provide new ideas for clinical treatment of IS.
Collapse
Affiliation(s)
- Hua Zhu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China.,Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yonggang Zhang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China.,Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yi Zhong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yingze Ye
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xinyao Hu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China.,Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
29
|
Belair DG, Lee JS, Kellner AV, Huard J, Murphy WL. Receptor mimicking TGF-β1 binding peptide for targeting TGF-β1 signaling. Biomater Sci 2021; 9:645-652. [PMID: 33289741 PMCID: PMC9254699 DOI: 10.1039/d0bm01374a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Prolonged and elevated transforming growth factor-β1 (TGF-β1) signaling can lead to undesired scar formation during tissue repair and fibrosis that is often a result of chronic inflammation in the lung, kidney, liver, heart, skin, and joints. We report new TGF-β1 binding peptides that interfere with TGF-β1 binding to its cognate receptors and thus attenuate its biological activity. We identified TGF-β1 binding peptides from the TGF-β1 binding domains of TGF-β receptors and engineered their sequences to facilitate chemical conjugation to biomaterials using molecular docking simulations. The in vitro binding studies and cell-based assays showed that RIPΔ, which was derived from TGF-β type I receptor, bound TGF-β1 in a sequence-specific manner and reduced the biological activity of TGF-β1 when the peptide was presented either in soluble form or conjugated to a commonly used synthetic biomaterial. This approach may have implications for clinical applications such as treatment of various fibrotic diseases and soft tissue repair and offer a design strategy for peptide antibodies based on the biomimicry of ligand-receptor interactions.
Collapse
Affiliation(s)
- David G Belair
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA.
| | - Jae Sung Lee
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, USA
| | - Anna V Kellner
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | - Johnny Huard
- Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, CO, USA
| | - William L Murphy
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA. and Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, USA and Materials Science Program, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
30
|
Hooglugt A, van der Stoel MM, Boon RA, Huveneers S. Endothelial YAP/TAZ Signaling in Angiogenesis and Tumor Vasculature. Front Oncol 2021; 10:612802. [PMID: 33614496 PMCID: PMC7890025 DOI: 10.3389/fonc.2020.612802] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/07/2020] [Indexed: 12/14/2022] Open
Abstract
Solid tumors are dependent on vascularization for their growth. The hypoxic, stiff, and pro-angiogenic tumor microenvironment induces angiogenesis, giving rise to an immature, proliferative, and permeable vasculature. The tumor vessels promote tumor metastasis and complicate delivery of anti-cancer therapies. In many types of tumors, YAP/TAZ activation is correlated with increased levels of angiogenesis. In addition, endothelial YAP/TAZ activation is important for the formation of new blood and lymphatic vessels during development. Oncogenic activation of YAP/TAZ in tumor cell growth and invasion has been studied in great detail, however the role of YAP/TAZ within the tumor endothelium remains insufficiently understood, which complicates therapeutic strategies aimed at targeting YAP/TAZ in cancer. Here, we overview the upstream signals from the tumor microenvironment that control endothelial YAP/TAZ activation and explore the role of their downstream targets in driving tumor angiogenesis. We further discuss the potential for anti-cancer treatments and vascular normalization strategies to improve tumor therapies.
Collapse
Affiliation(s)
- Aukie Hooglugt
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, VU University Medical Center, Amsterdam, Netherlands
| | - Miesje M. van der Stoel
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Reinier A. Boon
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, VU University Medical Center, Amsterdam, Netherlands
- German Center for Cardiovascular Research (DZHK), Partner Site Rhein-Main, Berlin, Germany
- Institute of Cardiovascular Regeneration, Goethe University, Frankfurt am Main, Germany
| | - Stephan Huveneers
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
31
|
Guo HF, Mohd Ali R, Abd Hamid R, Chang SK, Rahman MH, Zainal Z, Khaza'ai H. Epidermal Growth Factor and Tocotrienol-Rich Fraction Cream Formulation Accelerates Burn Healing Process Based on Its Gene Expression Pattern in Deep Partial-Thickness Burn Wound Model. INT J LOW EXTR WOUND 2020; 21:544-554. [PMID: 33241700 DOI: 10.1177/1534734620971066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Our previous study has demonstrated that epidermal growth factor (EGF) with tocotrienol-rich fraction (TRF) cream formulation accelerating postburn wound healing with deep partial-thickness burn in rats. Current study was conducted to determine the gene expression levels related to burn wound healing process. A total of 180 Sprague-Dawley rats were randomly divided into 6 groups: untreated control, treated with Silverdin cream, base cream, base cream with 0.00075% EGF, base cream with 3% TRF or base cream with 0.00075% EGF, and 3% TRF, respectively. Burn wounds were created and the above-mentioned creams were applied once daily. Six animals from each group were sacrificed on days 3, 7, 11, 14, and 21 postburn. RNA was extracted from wound tissues and quantitative real-time polymerase chain reaction was performed to analyze the 9 wound healing-related genes against time postburn. Results demonstrated that topically applied EGF + TRF formulation downregulated the expression levels of IL-6 (interluekin-6), TNF-α (tumor necrosis factor-α) and iNOS (inducible nitric oxide synthase) throughout the whole healing process. TGF-β1 (transforming growth factor-β) and VEGF-A (vascular endothelial growth factor-A) were reduced on day 14 postburn. On the contrary, increased expression of Collagen-1 in the early stage of wound healing was observed with no effects on epidemal growth factor receptor (EGFR). The results showed beneficial application of EGF + TRF cream in the treatment of burn wound since it accelerated wound healing by relieving oxidative stress, decreasing inflammation, and promoting proper tissue modelling in the burn wound.
Collapse
Affiliation(s)
- Hui-Fang Guo
- Chengde Medical University, Chengde Hebei, China.,Universiti Putra Malaysia, Serdang, Malaysia
| | | | | | - Sui Kiat Chang
- South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | | | - Zaida Zainal
- Malaysian Palm Oil Board, Bandar Baru Bangi, Kajang, Selangor, Malaysia
| | | |
Collapse
|
32
|
HOX Genes Family and Cancer: A Novel Role for Homeobox B9 in the Resistance to Anti-Angiogenic Therapies. Cancers (Basel) 2020; 12:cancers12113299. [PMID: 33171691 PMCID: PMC7695342 DOI: 10.3390/cancers12113299] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 10/30/2020] [Accepted: 11/06/2020] [Indexed: 01/05/2023] Open
Abstract
Simple Summary The inhibition of angiogenesis, relying on the use of drugs targeting the VEGF signaling pathway, has become one of the main strategies for cancer treatment. However, the intrinsic and acquired resistance to this type of therapy limit its efficacy. Thus, the identification of novel therapeutic targets is urgently needed. The resistance to anti-angiogenic treatment often occurs through the activation of alternative VEGF independent signaling pathways and recruitment of bone marrow-derived pro-angiogenic cells in the tumor microenvironment. HOX genes are key regulators of embryonic development, also involved in angiogenesis and in cancer progression. HOXB9 upregulation occurs in many types of cancer and it has been identified as a critical transcription factor involved in tumour resistance to anti-angiogenic drugs. Indeed, HOXB9 modulates the expression of alternative pro-angiogenic secreted factors in the tumour microenvironment leading tumor escape from the anti-angiogenic treatments. Hence, HOXB9 could serves as a novel therapeutic target to overcome the resistance to anti-angiogenic therapies. Abstract Angiogenesis is one of the hallmarks of cancer, and the inhibition of pro-angiogenic factors and or their receptors has become a primary strategy for cancer therapy. However, despite promising results in preclinical studies, the majority of patients either do not respond to these treatments or, after an initial period of response, they develop resistance to anti-angiogenic agents. Thus, the identification of a novel therapeutic target is urgently needed. Multiple mechanisms of resistance to anti-angiogenic therapy have been identified, including the upregulation of alternative angiogenic pathways and the recruitment of pro-angiogenic myeloid cells in the tumor microenvironment. Homeobox containing (HOX) genes are master regulators of embryonic development playing a pivotal role during both embryonic vasculogenesis and pathological angiogenesis in adults. The importance of HOX genes during cancer progression has been reported in many studies. In this review we will give a brief description of the HOX genes and their involvement in angiogenesis and cancer, with particular emphasis on HOXB9 as a possible novel target for anti-angiogenic therapy. HOXB9 upregulation has been reported in many types of cancers and it has been identified as a critical transcription factor involved in resistance to anti-angiogenic drugs.
Collapse
|
33
|
Kittipibul V, Vutthikraivit W, Kewcharoen J, Rattanawong P, Tantrachoti P, Putthapiban P, Nair N. Angiotensin II antagonists and gastrointestinal bleeding in left ventricular assist devices: A systematic review and meta-analysis. Int J Artif Organs 2020; 44:215-220. [PMID: 32842844 DOI: 10.1177/0391398820951811] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Gastrointestinal bleeding (GIB) especially from arteriovenous malformations (AVM) remains one of the devastating complications following continuous-flow left ventricular device (CF-LVAD) implantation. Blockade of angiotensin II pathway using angiotensin-converting enzyme inhibitors (ACEI)/angiotensin receptor blockers (ARB) was reported to mitigate the risk of GIB and AVM-related GIB by suppressing angiogenesis. We performed a systematic review and meta-analysis to evaluate the association between ACEI/ARB treatment and GIB in CF-LVAD population. Comprehensive literature search was performed through December 2019. We included studies reporting risk of GIB and/or AVM-related GIB events in LVAD patients who received ACEI/ARB with those who did not. Data from each study were combined using the random-effects to calculate odd ratios and 95% confidence intervals. Three retrospective cohort studies were included in this meta-analysis involving 619 LVADs patients (467 patients receiving ACEI/ARB). The use of ACEI/ARB was statistically associated with decreased incidence of overall GIB (pooled OR 0.35, 95% CI 0.22-0.56, I2 = 0.0%, p < 0.001). There was a non-significant trend toward lower risk for AVM-related GIB in patients who received ACEI/ARB (pooled OR 0.46, 95% CI 0.19-1.07, I2 = 51%, p = 0.07). Larger studies with specific definitions of ACEI/ARB use and GIB are warranted to accurately determine the potential non-hemodynamic benefits of ACEI/ARB in CF-LVAD patients.
Collapse
Affiliation(s)
- Veraprapas Kittipibul
- Department of Internal Medicine, University of Miami/Jackson Memorial Hospital, Miami, FL, USA
| | - Wasawat Vutthikraivit
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Jakrin Kewcharoen
- University of Hawaii Internal Medicine Residency Program, Honolulu, HI, USA
| | | | - Pakpoom Tantrachoti
- Department of Cardiology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | | | - Nandini Nair
- Department of Cardiology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| |
Collapse
|
34
|
Desjarlais M, Dussault S, Rivera JC, Chemtob S, Rivard A. MicroRNA Expression Profiling of Bone Marrow-Derived Proangiogenic Cells (PACs) in a Mouse Model of Hindlimb Ischemia: Modulation by Classical Cardiovascular Risk Factors. Front Genet 2020; 11:947. [PMID: 32973881 PMCID: PMC7472865 DOI: 10.3389/fgene.2020.00947] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 07/28/2020] [Indexed: 12/11/2022] Open
Abstract
Background Classical cardiovascular risk factors (CRFs) are associated with impaired angiogenic activities of bone marrow–derived proangiogenic cells (PACs) related to peripheral artery diseases (PADs) and ischemia-induced neovascularization. MicroRNAs (miRs) are key regulators of gene expression, and they are involved in the modulation of PAC function and PAC paracrine activity. However, the effects of CRFs on the modulation of miR expression in PACs are unknown. Aims and Methods We used a model of hindlimb ischemia and next-generation sequencing to perform a complete profiling of miRs in PACs isolated from the bone marrow of mice subjected to three models of CRFs: aging, smoking (SMK) and hypercholesterolemia (HC). Results Approximately 570 miRs were detected in PACs in the different CRF models. When excluding miRs with a very low expression level (<100 RPM), 40 to 61 miRs were found to be significantly modulated by aging, SMK, or HC. In each CRF condition, we identified downregulated proangiogenic miRs and upregulated antiangiogenic miRs that could contribute to explain PAC dysfunction. Interestingly, several miRs were similarly downregulated (e.g., miR-542-3p, miR-29) or upregulated (e.g., miR-501, miR-92a) in all CRF conditions. In silico approaches including Kyoto Encyclopedia of Genes and Genomes and cluster dendogram analyses identified predictive effects of these miRs on pathways having key roles in the modulation of angiogenesis and PAC function, including vascular endothelial growth factor signaling, extracellular matrix remodeling, PI3K/AKT/MAPK signaling, transforming growth factor beta (TGFb) pathway, p53, and cell cycle progression. Conclusion This study describes for the first time the effects of CRFs on the modulation of miR profile in PACs related to PAD and ischemia-induced neovascularization. We found that several angiogenesis-modulating miRs are similarly altered in different CRF conditions. Our findings constitute a solid framework for the identification of miRs that could be targeted in PACs in order to improve their angiogenic function and for the future development of novel therapies to improve neovascularization and reduce tissue damage in patients with severe PAD.
Collapse
Affiliation(s)
- Michel Desjarlais
- Department of Medicine, Centre Hospitalier de l'Université de Montréal (CHUM) Research Center, Montréal, QC, Canada.,Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Center, University of Montréal, Montréal, QC, Canada
| | - Sylvie Dussault
- Department of Medicine, Centre Hospitalier de l'Université de Montréal (CHUM) Research Center, Montréal, QC, Canada
| | - José Carlos Rivera
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Center, University of Montréal, Montréal, QC, Canada.,Departments of Pediatrics, Ophthalmology and Pharmacology, Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, QC, Canada
| | - Sylvain Chemtob
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Center, University of Montréal, Montréal, QC, Canada.,Departments of Pediatrics, Ophthalmology and Pharmacology, Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, QC, Canada
| | - Alain Rivard
- Department of Medicine, Centre Hospitalier de l'Université de Montréal (CHUM) Research Center, Montréal, QC, Canada
| |
Collapse
|
35
|
Choudhary A, Kant V, Jangir BL, Joshi VG. Quercetin loaded chitosan tripolyphosphate nanoparticles accelerated cutaneous wound healing in Wistar rats. Eur J Pharmacol 2020; 880:173172. [PMID: 32407724 DOI: 10.1016/j.ejphar.2020.173172] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 04/30/2020] [Accepted: 05/04/2020] [Indexed: 12/25/2022]
Abstract
Previous studies have shown that quercetin on topical application improved cutaneous wound healing in rats, but hydrophobic nature and less skin penetration limits its potential as topical healing agent. Therefore, present study was planned to investigate wound healing potential of chitosan based quercetin nanoparticles. Quercetin loaded nanoparticles were synthesized by ionic gelation method and characterized by various standard techniques. A 2 × 2 cm2 square shaped wound was created on the thoraco-lumbar part of rats. Wounded rats were divided into 6 groups namely, gel (20%), blank nanoparticle (0.16%), bulk quercetin (0.3%), quercetin nanoparticles (0.03%), quercetin nanoparticles (0.1%) and quercetin nanoparticles (0.3%) treated groups. Different formulations of quercetin nanoparticles were applied on the wounds for the duration of study and healing tissues were collected on 7th, 14th and 21st day to study various parameters. Quercetin loaded nanoparticles were 361.16 ± 9.72 nm size and spherical in shape. We observed quercetin nanoparticles (0.03%) treatment caused marked reduction in the tumor necrosis factor-alpha, whereas expressions of interleukin 10, vascular endothelial growth factor and transforming growth factor beta1 was increased significantly with treatment. The granulation tissue of quercetin nanoparticles (0.03%) treated group showed better quality healing and maturity as supported by the increased blood vessels density, decreased inflammatory cells, increased number of myofibroblasts, deposition and arrangement of collagen fibers and re-epithelialization. In conclusion, quercetin nanoparticles (0.03%) treatment significantly improved wound healing by modulation of cytokines and growth factors involved in inflammatory and proliferative phases of wound healing.
Collapse
Affiliation(s)
- Abhinav Choudhary
- Department of Veterinary Pharmacology & Toxicology, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, Haryana, India
| | - Vinay Kant
- Department of Veterinary Pharmacology & Toxicology, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, Haryana, India.
| | - Babu Lal Jangir
- Department of Veterinary Pathology, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, Haryana, India
| | - V G Joshi
- Department of Animal Biotechnology, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, Haryana, India
| |
Collapse
|
36
|
Sakamoto R, Kajihara I, Miyauchi H, Maeda-Otsuka S, Yamada-Kanazawa S, Sawamura S, Kanemaru H, Makino K, Aoi J, Makino T, Fukushima S, Masuzawa M, Masuzawa M, Amoh Y, Hoshina D, Abe R, Ihn H. Inhibition of Endoglin Exerts Antitumor Effects through the Regulation of Non-Smad TGF-β Signaling in Angiosarcoma. J Invest Dermatol 2020; 140:2060-2072.e6. [PMID: 32142796 DOI: 10.1016/j.jid.2020.01.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 01/10/2020] [Accepted: 01/30/2020] [Indexed: 12/21/2022]
Abstract
Angiosarcoma is a rare malignant tumor derived from endothelial cells, and its prognosis is poor because advanced angiosarcoma is often resistant to taxane therapy. Endoglin (CD105) acts as a coreceptor for TGF-β signaling and is overexpressed in tumor-associated endothelial cells and enhances tumor angiogenesis. Numerous clinical trials are testing the effectiveness of anti-endoglin antibodies in various types of malignancies. Here, we investigated the role of endoglin in the pathogenesis of angiosarcoma and whether endoglin inhibition results in antitumor activity. Endoglin was overexpressed in angiosarcoma, and its inhibition was effective in promoting apoptosis and the suppression of migration, invasion, tube formation, and Warburg effect in angiosarcoma cells. Knockdown of endoglin activated caspase 3/7 that is essential for apoptosis, reduced survivin levels, and decreased paxillin and vascular endothelial cadherin phosphorylation and matrix metalloproteinase 2 and matrix metalloproteinase 9 activities in angiosarcoma cells. Although endoglin is a coreceptor that regulates TGF-β signaling, the antitumor effect of endoglin in angiosarcoma was not based on Smad signaling regulation but on non-Smad TGF-β signaling. Taken together, these results indicated that endoglin could be a novel therapeutic target for angiosarcoma.
Collapse
Affiliation(s)
- Ryoko Sakamoto
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Ikko Kajihara
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.
| | - Hitomi Miyauchi
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Saki Maeda-Otsuka
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Saori Yamada-Kanazawa
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Soichiro Sawamura
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hisashi Kanemaru
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Katsunari Makino
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Jun Aoi
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Takamitsu Makino
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Satoshi Fukushima
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Mamiko Masuzawa
- Department of Dermatology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Mikio Masuzawa
- Department of Molecular Diagnostics, School of Allied Health Sciences, Kitasato University, Kanagawa, Japan
| | - Yasuyuki Amoh
- Department of Dermatology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Daichi Hoshina
- Department of Dermatology, Hokkaido University Graduate School of Medicine, Hokkaido, Japan
| | - Riichiro Abe
- Department of Dermatology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Hironobu Ihn
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
37
|
Converse MP, Sobhanian M, Taber DJ, Houston BA, Meadows HB, Uber WE. Effect of Angiotensin II Inhibitors on Gastrointestinal Bleeding in Patients With Left Ventricular Assist Devices. J Am Coll Cardiol 2020; 73:1769-1778. [PMID: 30975293 DOI: 10.1016/j.jacc.2019.01.051] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 01/02/2019] [Accepted: 01/03/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Angiotensin II receptor activation may result in angiogenesis, and ultimately arteriovenous malformations (AVM), through transforming growth factor (TGF)-β and angiopoietin-2 pathway activation. OBJECTIVES The goal of this study was to determine whether angiotensin-converting enzyme (ACE) inhibitors or angiotensin receptor blockers (ARB) were associated with lower risk of major gastrointestinal bleeds (GIB) and AVM-related GIBs in continuous-flow left ventricular assist device (CF-LVAD) patients. METHODS The authors reviewed HeartMate II CF-LVAD recipients between January 2009 and July 2016. Major GIBs were endoscopically confirmed requiring ≥2 U of packed red blood cells or resulting in death. ACE inhibitor/ARB dose was abstracted from medical records. ACE inhibitor/ARB exposure status was landmarked at 30 days post-operatively to avoid immortal time bias. Fine and Gray hazard models assessed the impact of ACE inhibitor/ARB therapy on major GIB and AVM-related GIB, whereas standard Cox regression assessed the impact on mortality, adjusting for baseline variables. RESULTS One-hundred and eleven patients were included with a mean 2.1 ± 1.4 years follow-up. Patients who received an ACE inhibitor/ARB within 30 days post-operatively had a 57% reduction in the risk of major GIB (adjusted hazard ratio [aHR]: 0.43; 95% confidence interval [CI]: 0.19 to 0.97; p = 0.042) and a 63% reduction in the risk of AVM-related GIB (aHR: 0.37; 95% CI: 0.16 to 0.84; p = 0.017). When the mean daily post-operative lisinopril-equivalent ACE inhibitor/ARB dose was >5 mg, the risk of major GIB decreased in a dose-threshold manner (aHR: 0.28; 95% CI: 0.09 to 0.85; p = 0.025). CONCLUSIONS ACE inhibitor/ARB therapy is associated with a protective effect of developing GIBs in CF-LVAD patients, with a dose threshold of >5 mg of daily lisinopril equivalence, possibly due to prevention of AVM formation.
Collapse
Affiliation(s)
- Maureen P Converse
- Department of Pharmacy Services, University of Florida Health Shands Hospital, Gainesville, Florida.
| | - Minoosh Sobhanian
- Department of Pharmacy Services, Memorial Hermann Hospital-Texas Medical Center, Houston, Texas
| | - David J Taber
- Department of Pharmacy Services, Medical University of South Carolina, Charleston, South Carolina
| | - Brian A Houston
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Holly B Meadows
- Department of Pharmacy Services, Medical University of South Carolina, Charleston, South Carolina
| | - Walter E Uber
- Department of Pharmacy Services, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
38
|
The Predicament of Gastrointestinal Bleeding in Patients With a Continuous-Flow Left Ventricular Assist Device: Pathophysiology, Evaluation, and Management. Cardiol Rev 2020; 27:222-229. [PMID: 30365405 DOI: 10.1097/crd.0000000000000235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Heart failure affects over 5 million Americans, with numbers expected to rise. While heart transplantation is the most effective long-term strategy for end-stage heart failure, there is a limited cardiac donor pool, and these organs are often unavailable at the time of need. Left ventricular assist devices, therefore, continue to be used to bridge this gap. Originally implanted as a bridge to transplant, these devices are now additionally utilized as destination therapy for patients ineligible for transplant. With the widespread applicability of these devices for not just temporary measures, but also for prolonged use, the short- and long-term impact on other organ systems has become more evident. For example, gastrointestinal (GI) bleeding, with an incidence approaching 30%, is one such complication post-continuous-flow left ventricular assist device implantation. This high incidence of GI bleeding is thought to stem from a combination of factors, including the need for concomitant anticoagulant and antiplatelet therapy, and intrinsic device-related properties resulting in acquired Von Willebrand disease and arteriovenous malformations. Due to the significant morbidity associated with these GI bleeding events, a standardized protocol optimizing medical and endoscopic management, alongside close coordination between the gastroenterology and cardiology services, should be advocated for and ultimately employed.
Collapse
|
39
|
Moradi-Marjaneh R, Khazaei M, Ferns GA, Aghaee-Bakhtiari SH. The Role of TGF-β Signaling Regulatory MicroRNAs in the Pathogenesis of Colorectal Cancer. Curr Pharm Des 2019; 24:4611-4618. [PMID: 30636580 DOI: 10.2174/1381612825666190110150705] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 12/24/2018] [Accepted: 12/31/2018] [Indexed: 02/08/2023]
Abstract
Colorectal cancer (CRC) is one of the most common cancers globally and is associated with a high mortality rate. The transforming growth factor beta (TGF-β) signaling pathway plays an important role in normal intestinal tissue function, but has also been implicated in the development of CRC. MicroRNAs (miRNAs) have also recently emerged as important regulators of cancer development and progression. They act by targeting multiple signaling pathways including the TGF-β signaling pathway. There is growing evidence demonstrating that miRNAs target various components of the TGF-β signaling pathway, including TGF-β1, TGF-β2, regulatory SMADs (SMAD1, 2, 3, 5 and 9), co-mediator SMAD4, inhibitory SMADs (SMAD6 and 7) and the TGF-β receptors, and thereby alter the proliferation and migration of CRC cells. In this review, we summarize the data concerning the interaction between TGF-β signaling pathway and miRNAs with the aim to better understanding the CRC molecular mechanisms and hence better management of this disease.
Collapse
Affiliation(s)
- Reyhaneh Moradi-Marjaneh
- Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran.,Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex BN1 9PH, United Kingdom
| | - Seyed H Aghaee-Bakhtiari
- Bioinformatics Research Group, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
40
|
Akla N, Viallard C, Popovic N, Lora Gil C, Sapieha P, Larrivée B. BMP9 (Bone Morphogenetic Protein-9)/Alk1 (Activin-Like Kinase Receptor Type I) Signaling Prevents Hyperglycemia-Induced Vascular Permeability. Arterioscler Thromb Vasc Biol 2019; 38:1821-1836. [PMID: 29880487 DOI: 10.1161/atvbaha.118.310733] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Objective- Diabetic macular edema is a major cause of visual impairment. It is caused by blood-retinal barrier breakdown that leads to vascular hyperpermeability. Current therapeutic approaches consist of retinal photocoagulation or targeting VEGF (vascular endothelial growth factor) to limit vascular leakage. However, long-term intravitreal use of anti-VEGFs is associated with potential safety issues, and the identification of alternative regulators of vascular permeability may provide safer therapeutic options. The vascular specific BMP (bone morphogenetic protein) receptor ALK1 (activin-like kinase receptor type I) and its circulating ligand BMP9 have been shown to be potent vascular quiescence factors, but their role in the context of microvascular permeability associated with hyperglycemia has not been evaluated. Approach and Results- We investigated Alk1 signaling in hyperglycemic endothelial cells and assessed whether BMP9/Alk1 signaling could modulate vascular permeability. We show that high glucose concentrations impair Alk1 signaling, both in cultured endothelial cells and in a streptozotocin model of mouse diabetes mellitus. We observed that Alk1 signaling participates in the maintenance of vascular barrier function, as Alk1 haploinsufficiency worsens the vascular leakage observed in diabetic mice. Conversely, sustained delivery of BMP9 by adenoviral vectors significantly decreased the loss of retinal barrier function in diabetic mice. Mechanistically, we demonstrate that Alk1 signaling prevents VEGF-induced phosphorylation of VE-cadherin and induces the expression of occludin, thus strengthening vascular barrier functions. Conclusions- From these data, we suggest that by preventing retinal vascular permeability, BMP9 could serve as a novel therapeutic agent for diabetic macular edema.
Collapse
Affiliation(s)
- Naoufal Akla
- From the Department of Biochemistry (N.A., P.S.).,University of Montreal, Quebec, Canada; and Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec, Canada (N.A., C.V., N.P., C.L.G., P.S., B.L.)
| | - Claire Viallard
- Department of Molecular Biology (C.V., B.L.).,University of Montreal, Quebec, Canada; and Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec, Canada (N.A., C.V., N.P., C.L.G., P.S., B.L.)
| | - Natalija Popovic
- Department of Biomedical Sciences (N.P., C.L.G., B.L.).,University of Montreal, Quebec, Canada; and Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec, Canada (N.A., C.V., N.P., C.L.G., P.S., B.L.)
| | - Cindy Lora Gil
- Department of Biomedical Sciences (N.P., C.L.G., B.L.).,University of Montreal, Quebec, Canada; and Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec, Canada (N.A., C.V., N.P., C.L.G., P.S., B.L.)
| | - Przemyslaw Sapieha
- From the Department of Biochemistry (N.A., P.S.).,Department of Ophthalmology (P.S., B.L.).,University of Montreal, Quebec, Canada; and Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec, Canada (N.A., C.V., N.P., C.L.G., P.S., B.L.)
| | - Bruno Larrivée
- Department of Molecular Biology (C.V., B.L.).,Department of Biomedical Sciences (N.P., C.L.G., B.L.).,Department of Ophthalmology (P.S., B.L.).,University of Montreal, Quebec, Canada; and Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec, Canada (N.A., C.V., N.P., C.L.G., P.S., B.L.)
| |
Collapse
|
41
|
Min E, Schwartz MA. Translocating transcription factors in fluid shear stress-mediated vascular remodeling and disease. Exp Cell Res 2019; 376:92-97. [PMID: 30633880 DOI: 10.1016/j.yexcr.2019.01.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 01/04/2019] [Accepted: 01/07/2019] [Indexed: 02/06/2023]
Abstract
Endothelial cells are exposed to fluid shear stress profiles that vary in magnitude, pulsatility, and directionality due to regional variations in blood vessel structure. Laminar flow at physiological levels is atheroprotective; multidirectional or reversing low (disturbed) flow promotes inflammation and disease; and high or low laminar flow promote outward or inward remodeling, respectively. However, our understanding of how endothelial cells discern these different flow profiles and regulate gene expression accordingly is limited. This article reviews recent studies that identify the TGFβ/Smad, Notch, Yap/Taz, and Wnt/β-catenin pathways as important mediators of flow profile- and magnitude-dependent signaling.
Collapse
Affiliation(s)
- Elizabeth Min
- Department of Cell Biology, Yale School of Medicine, United States; Yale Cardiovascular Research Center, United States
| | - Martin A Schwartz
- Department of Cell Biology, Yale School of Medicine, United States; Yale Cardiovascular Research Center, United States; Department of Medicine (Cardiology), United States; Department of Biomedical Engineering, United States.
| |
Collapse
|
42
|
Natividad-Diaz SL, Browne S, Jha AK, Ma Z, Hossainy S, Kurokawa YK, George SC, Healy KE. A combined hiPSC-derived endothelial cell and in vitro microfluidic platform for assessing biomaterial-based angiogenesis. Biomaterials 2018; 194:73-83. [PMID: 30583150 DOI: 10.1016/j.biomaterials.2018.11.032] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 11/26/2018] [Indexed: 12/12/2022]
Abstract
Human induced pluripotent stem cell (hiPSC) derived angiogenesis models present a unique opportunity for patient-specific platforms to study the complex process of angiogenesis and the endothelial cell response to biomaterial and biophysical changes in a defined microenvironment. We present a refined method for differentiating hiPSCs into a CD31 + endothelial cell population (hiPSC-ECs) using a single basal medium from pluripotency to the final stage of differentiation. This protocol produces endothelial cells that are functionally competent in assays following purification. Subsequently, an in vitro angiogenesis model was developed by encapsulating the hiPSC-ECs into a tunable, growth factor sequestering hyaluronic acid (HyA) matrix where they formed stable, capillary-like networks that responded to environmental stimuli. Perfusion of the networks was demonstrated using fluorescent beads in a microfluidic device designed to study angiogenesis. The combination of hiPSC-ECs, bioinspired hydrogel, and the microfluidic platform creates a unique testbed for rapidly assessing the performance of angiogenic biomaterials.
Collapse
Affiliation(s)
- Sylvia L Natividad-Diaz
- Department of Bioengineering, University of California-Berkeley, Berkeley, CA, United States
| | - Shane Browne
- Department of Bioengineering, University of California-Berkeley, Berkeley, CA, United States
| | - Amit K Jha
- Department of Bioengineering, University of California-Berkeley, Berkeley, CA, United States; Department of Materials Science and Engineering, University of California-Berkeley, Berkeley, CA, United States
| | - Zhen Ma
- Department of Bioengineering, University of California-Berkeley, Berkeley, CA, United States
| | - Samir Hossainy
- Department of Bioengineering, University of California-Berkeley, Berkeley, CA, United States; Department of Materials Science and Engineering, University of California-Berkeley, Berkeley, CA, United States
| | - Yosuke K Kurokawa
- Department of Biomedical Engineering, Washington University in St Louis, MO, United States
| | - Steven C George
- Department of Biomedical Engineering, University of California-Davis, Davis, CA, United States
| | - Kevin E Healy
- Department of Bioengineering, University of California-Berkeley, Berkeley, CA, United States; Department of Materials Science and Engineering, University of California-Berkeley, Berkeley, CA, United States.
| |
Collapse
|
43
|
Griessenauer CJ, Farrell S, Sarkar A, Zand R, Abedi V, Holland N, Michael A, Cummings CL, Metpally R, Carey DJ, Goren O, Martin N, Hendrix P, Schirmer CM. Genetic susceptibility to cerebrovascular disease: A systematic review. J Cereb Blood Flow Metab 2018; 38:1853-1871. [PMID: 30182779 PMCID: PMC6259318 DOI: 10.1177/0271678x18797958] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Investigation of genetic susceptibility to cerebrovascular disease has been of growing interest. A systematic review of human studies assessing neurogenomic aspects of cerebrovascular disease was performed according to the preferred reporting items for systematic reviews and meta-analyses (PRISMA) statement. Any association study exploring genetic variants located in the exome associated with one of the major cerebrovascular diseases with at least 500 subjects was eligible for inclusion. Of 6874 manuscripts identified, 35 studies met the inclusion criteria. Most studies of interest focused on ischemic stroke and cerebrovascular occlusive disease. Large cohort genetic association studies on hemorrhagic cerebrovascular disease were less common. In addition to rare, well-established monogenic conditions with significant risk for cerebrovascular disease, a number of genetic variants are also relevant to cerebrovascular pathogenesis as part of a multifactorial process. The 45 polymorphisms identified were located in genes involved in processes related to endothelial and vascular health (15 (33.4%) variants), plasma lipid metabolism (10 (22.2%) variants), inflammation (9 (20%) variants), coagulation (3 (6.7%) variants), and blood pressure modulation (2 (4.4%) variants), and other (6 (13.3%) variants). This work represents a comprehensive overview of genetic variants in the exome relevant to ischemic and hemorrhagic stroke pathophysiology.
Collapse
Affiliation(s)
- Christoph J Griessenauer
- 1 Department of Neurosurgery, Geisinger Commonwealth School of Medicine, Geisinger, Danville, PA, USA.,2 Research Institute of Neurointervention, Paracelsus Medical University, Salzburg, Austria
| | - Sean Farrell
- 3 Geisinger Commonwealth School of Medicine, Scranton, PA, USA
| | - Atom Sarkar
- 1 Department of Neurosurgery, Geisinger Commonwealth School of Medicine, Geisinger, Danville, PA, USA
| | - Ramin Zand
- 4 Department of Neurology, Geisinger Commonwealth School of Medicine, Geisinger, Danville, PA, USA
| | - Vida Abedi
- 5 Biomedical and Translational Informatics Institute, Geisinger, Danville, PA, USA
| | - Neil Holland
- 4 Department of Neurology, Geisinger Commonwealth School of Medicine, Geisinger, Danville, PA, USA
| | - Andrew Michael
- 6 Neuroimaging Analytics Laboratory, Autism & Developmental Medicine Institute, Geisinger, Lewisburg, PA, USA
| | - Christopher L Cummings
- 4 Department of Neurology, Geisinger Commonwealth School of Medicine, Geisinger, Danville, PA, USA
| | | | | | - Oded Goren
- 1 Department of Neurosurgery, Geisinger Commonwealth School of Medicine, Geisinger, Danville, PA, USA
| | - Neil Martin
- 1 Department of Neurosurgery, Geisinger Commonwealth School of Medicine, Geisinger, Danville, PA, USA
| | - Philipp Hendrix
- 8 Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg/Saar, Germany
| | - Clemens M Schirmer
- 1 Department of Neurosurgery, Geisinger Commonwealth School of Medicine, Geisinger, Danville, PA, USA
| |
Collapse
|
44
|
Ijaz HM, Lodhi MU, Chowdhury W, Syed IA, Patel C, McDaniel BA, Rahim M. Co-existing Gastrointestinal Hemorrhage and Deep Vein Thrombosis in a Patient with Hereditary Hemorrhagic Telangiectasia: Management Dilemma. Cureus 2018; 10:e3305. [PMID: 30949422 PMCID: PMC6436667 DOI: 10.7759/cureus.3305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Hereditary hemorrhagic telangiectasia (HHT) is described as a vascular defect, causing recurrent visceral and mucocutaneous bleeding. It is an autosomal dominant disease and has variable expressivity. The phenotypic presentation is dependent on the type of gene defect. Recurrent epistaxis is the most common symptom, along with gastrointestinal (GI), pulmonary, and arteriovenous malformations (AVM). The Curacao criteria are used to make the diagnosis of HHT. Genetic sequence testing for endoglin (ENG) or activin receptor-like kinase type 1 (ALK1) can be performed to confirm the diagnosis. However, genetic sequencing is not necessary. Along with recurrent bleeding, patients with HHT also have an increased risk of thromboembolic events. Supportive treatment prevents acute symptoms, but the therapeutic options of HHT are based on multiple factors. We describe the case of a 69-year-old male who presented with GI bleeding and a history of HHT and recurrent deep vein thrombosis (DVT). We discuss the diagnostic guidelines and treatment options for patients with HHT. Furthermore, we also discuss the challenge in treating patients with co-existing GI bleeding and DVT, as in our case.
Collapse
Affiliation(s)
- Hasnan M Ijaz
- Internal Medicine, Raleigh General Hospital, Beckley, USA
| | | | | | | | - Chirag Patel
- Internal Medicine, Charleston Area Medical Center / West Virginia University, Charleston, USA
| | - Bryce A McDaniel
- Internal Medicine, Charleston Area Medical Center / West Virginia University, Charleston, USA
| | - Mustafa Rahim
- Internal Medicine, West Virginia University School of Medicine, Morgantown, USA
| |
Collapse
|
45
|
|
46
|
Singh P, Jenkins LM, Horst B, Alers V, Pradhan S, Kaur P, Srivastava T, Hempel N, Győrffy B, Broude EV, Lee NY, Mythreye K. Inhibin Is a Novel Paracrine Factor for Tumor Angiogenesis and Metastasis. Cancer Res 2018; 78:2978-2989. [PMID: 29535220 PMCID: PMC6510404 DOI: 10.1158/0008-5472.can-17-2316] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 01/03/2018] [Accepted: 03/09/2018] [Indexed: 01/21/2023]
Abstract
Inhibin is a heterodimeric TGFβ family ligand that is expressed in many cancers and is a selective biomarker for ovarian cancers; however, its tumor-specific functions remain unknown. Here, we demonstrate that the α subunit of inhibin (INHA), which is critical for the functionality of dimeric inhibin A/B, correlates with microvessel density in human ovarian tissues and is predictive of poor clinical outcomes in multiple cancers. We demonstrate that inhibin-regulated angiogenesis is necessary for metastasis. Although inhibin had no direct impact on tumor cell signaling, both tumor cell-derived and recombinant inhibin elicit a strong paracrine response from endothelial cells by triggering SMAD1/5 activation and angiogenesis in vitro and in vivo Inhibin-induced angiogenesis was abrogated via anti-inhibin α antibodies. The endothelial-specific TGFβ receptor complex comprising ALK1 and endoglin was a crucial mediator of inhibin signaling, offering a molecular mechanism for inhibin-mediated angiogenesis. These results are the first to define a role for inhibin in tumor metastasis and vascularization and offer an antibody-based approach for targeting inhibin therapeutically.Significance: Inhibin is a predictor of poor patient survival in multiple cancers and is a potential target for antiangiogenic therapies. Cancer Res; 78(11); 2978-89. ©2018 AACR.
Collapse
Affiliation(s)
- Priyanka Singh
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina
| | - Laura M Jenkins
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina
| | - Ben Horst
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina
| | - Victoria Alers
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina
| | - Shrikant Pradhan
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina
| | - Prabhjot Kaur
- Department of Genetics, University of Delhi, South Campus, India
| | | | - Nadine Hempel
- Department of Pharmacology, Penn State University College of Medicine, Hershey, Pennsylvania
| | - Balázs Győrffy
- MTA TTK Lendület Cancer Biomarker Research Group, Institute of Enzymology, and Semmelweis University 2nd Department of Pediatrics, Budapest, Hungary
| | - Eugenia V Broude
- Department of Drug Discovery and Biomedical Sciences, School of Pharmacy, Ohio State University, Columbus, Ohio
| | - Nam Y Lee
- Division of Pharmacology, College of Pharmacy, Ohio State University, Columbus, Ohio
| | - Karthikeyan Mythreye
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina.
- Department of Drug Discovery and Biomedical Sciences, School of Pharmacy, Ohio State University, Columbus, Ohio
| |
Collapse
|
47
|
Verrecchia F, Rédini F. Transforming Growth Factor-β Signaling Plays a Pivotal Role in the Interplay Between Osteosarcoma Cells and Their Microenvironment. Front Oncol 2018; 8:133. [PMID: 29761075 PMCID: PMC5937053 DOI: 10.3389/fonc.2018.00133] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 04/13/2018] [Indexed: 12/12/2022] Open
Abstract
Osteosarcomas are the most frequent form of primary bone tumors and mainly affect children, adolescents, and young adults. Despite encouraging progress in therapeutic management, including the advent of multidrug chemotherapy, the survival rates have remained unchanged for more than four decades: 75% at 5 years for localized disease, but two groups of patients are still at high risk: metastatic at diagnosis (overall survival around 40% at 5 years) and/or poor responders to chemotherapy (20% at 5 years). Because these tumors are classified as “complex genomic,” it is extremely difficult to determine the signaling pathways that might be targeted by specific therapies. A hypothesis has thus emerged, stating that the particular microenvironment of these tumors may interfere with the tumor cells that promote chemoresistance and the dissemination of metastases. The stroma is composed of a large number of cell types (immune cells, endothelial cells, mesenchymal stromal cells, etc.) which secrete growth factors, such as transforming growth factor-β (TGF-β), which favors the development of primary tumors and dissemination of metastases by constituting a permissive niche at primary and distant sites. Rather than targeting the tumor cells themselves, which are very heterogeneous in osteosarcoma, the hypothesis is instead to target the key actors secreted in the microenvironment, such as TGF-βs, which play a part in tumor progression. In the last decade, numerous studies have shown that overexpression of TGF-β is a hallmark of many cancers, including primary bone tumors. In this context, TGF-β signaling has emerged as a crucial factor in the cross talk between tumor cells and stroma cells in poor-prognosis cancers. Secretion of TGF-β by tumor cells or stroma cells can effectively act in a paracrine manner to regulate the phenotype and functions of the microenvironment to stimulate protumorigenic microenvironmental changes. TGF-β can thus exert its protumorigenic function in primary bone tumors by promoting angiogenesis, bone remodeling and cell migration, and by inhibiting immunosurveillance. This review focuses on the involvement of TGF-β signaling in primary bone tumor development, and the related therapeutic options that may be possible for these tumors.
Collapse
Affiliation(s)
- Franck Verrecchia
- UMR1238 INSERM, Université de Nantes, PHY-OS, "Bone Sarcomas and Remodeling of Calcified Tissues", Medical School, Nantes, France
| | - Françoise Rédini
- UMR1238 INSERM, Université de Nantes, PHY-OS, "Bone Sarcomas and Remodeling of Calcified Tissues", Medical School, Nantes, France
| |
Collapse
|
48
|
"Vessels in the Storm": Searching for Prognostic and Predictive Angiogenic Factors in Colorectal Cancer. Int J Mol Sci 2018; 19:ijms19010299. [PMID: 29351242 PMCID: PMC5796244 DOI: 10.3390/ijms19010299] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 01/08/2018] [Accepted: 01/11/2018] [Indexed: 12/22/2022] Open
Abstract
High expectations are placed upon anti-angiogenic compounds for metastatic colorectal cancer (mCRC), the first malignancy for which such type of treatment has been approved. Indeed, clinical trials have confirmed that targeting the formation of new vessels can improve in many cases clinical outcomes of mCRC patients. However, current anti-angiogenic drugs are far from obtaining the desirable or expected curative results. Many are the factors probably involved in such disappointing results, but particular attention is currently focused on the validation of biomarkers able to improve the direction of treatment protocols. Because clinical studies have clearly demonstrated that serum or tissue concentration of some angiogenic factors is associated with the evolution of the disease of mCRC patients, they are currently explored as potential biomarkers of prognosis and of tumor response to therapy. However, the complex biology underlying CRC -induced angiogenesis is a hurdle in finding rapid solutions. The aim of this review was to explore molecular mechanisms that determine the formation of tumor-associated vessels during CRC progression, and to discuss the potential role of angiogenic factors as diagnostic, prognostic and predictive biomarkers in CRC.
Collapse
|
49
|
|
50
|
Chruścik A, Gopalan V, Lam AKY. The clinical and biological roles of transforming growth factor beta in colon cancer stem cells: A systematic review. Eur J Cell Biol 2018; 97:15-22. [PMID: 29128131 DOI: 10.1016/j.ejcb.2017.11.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 11/07/2017] [Accepted: 11/07/2017] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Transforming growth factor beta (TGF-β) is a multipurpose cytokine, which plays a role in many cellular functions such as proliferation, differentiation, migration, apoptosis, cell adhesion and regulation of epithelial to mesenchymal transition. Despite many studies having observed the effect that TGF-β plays in colorectal cancer, its role in the colorectal stem cell population has not been widely observed. METHOD This systematic review will analyse the role of TGF-β in the stem cell population of colorectal cancer. RESULTS The effects on the stem cell phenotype are through the downstream proteins involved in activation of the TGF-β pathway. Its involvement in the initiation of the epithelial to mesenchymal transition (EMT), the effect of colorectal invasion and metastasis regulated through the Smad protein involvement in the EMT, initiation of angiogenesis, promotion of metastasis of colorectal cancer to the liver and its ability to cross-talk with other pathways. CONCLUSION TGF-β is a key player in angiogenesis, tumour growth and metastasis in colon cancer.
Collapse
Affiliation(s)
- Anna Chruścik
- Cancer Molecular Pathology, School of Medicine and Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia
| | - Vinod Gopalan
- Cancer Molecular Pathology, School of Medicine and Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia
| | - Alfred King-Yin Lam
- Cancer Molecular Pathology, School of Medicine and Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia.
| |
Collapse
|