151
|
Ashar-Patel A, Kaymaz Y, Rajakumar A, Bailey JA, Karumanchi SA, Moore MJ. FLT1 and transcriptome-wide polyadenylation site (PAS) analysis in preeclampsia. Sci Rep 2017; 7:12139. [PMID: 28939845 PMCID: PMC5610261 DOI: 10.1038/s41598-017-11639-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 08/25/2017] [Indexed: 12/18/2022] Open
Abstract
Maternal symptoms of preeclampsia (PE) are primarily driven by excess anti-angiogenic factors originating from the placenta. Chief among these are soluble Flt1 proteins (sFlt1s) produced from alternatively polyadenylated mRNA isoforms. Here we used polyadenylation site sequencing (PAS-Seq) of RNA from normal and PE human placentae to interrogate transcriptome-wide gene expression and alternative polyadenylation signatures associated with early-onset PE (EO-PE; symptom onset < 34 weeks) and late-onset PE (LO-PE; symptom onset > 34 weeks) cohorts. While we observed no general shift in alternative polyadenylation associated with PE, the EO-PE and LO-PE cohorts do exhibit gene expression profiles distinct from both each other and from normal placentae. The only two genes upregulated across all transcriptome-wide PE analyses to date (microarray, RNA-Seq and PAS-Seq) are NRIP1 (RIP140), a transcriptional co-regulator linked to metabolic syndromes associated with obesity, and Flt1. Consistent with sFlt1 overproduction being a significant driver of clinical symptoms, placental Flt1 mRNA levels strongly correlate with maternal blood pressure. For Flt1, just three mRNA isoforms account for > 94% of all transcripts, with increased transcription of the entire locus driving Flt1 upregulation in both EO-PE and LO-PE. These three isoforms thus represent potential targets for therapeutic RNA interference (RNAi) in both early and late presentations.
Collapse
Affiliation(s)
- Ami Ashar-Patel
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, USA
| | - Yasin Kaymaz
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Boston, MA, USA
| | - Augustine Rajakumar
- Departments of Gynecology and Obstetrics, Emory University, Atlanta, USA.,Departments of Medicine, Obstetrics and Gynecology and Center for Vascular Biology Research, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Jeffrey A Bailey
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Boston, MA, USA.,Division of Transfusion Medicine, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - S Ananth Karumanchi
- Departments of Medicine, Obstetrics and Gynecology and Center for Vascular Biology Research, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Melissa J Moore
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
152
|
Li M, Kroetz DL. Bevacizumab-induced hypertension: Clinical presentation and molecular understanding. Pharmacol Ther 2017; 182:152-160. [PMID: 28882537 DOI: 10.1016/j.pharmthera.2017.08.012] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Bevacizumab is a vascular endothelial growth factor-A-specific angiogenesis inhibitor indicated as an adjunct to chemotherapy for the treatment of several types of cancer. Hypertension is commonly observed during bevacizumab treatment, and high-grade toxicity can limit therapy and lead to other cardiovascular complications. The factors that contribute to interindividual variability in blood pressure response to bevacizumab treatment are not well understood. In this review, we outline research efforts to understand the mechanisms and pathophysiology of hypertension resulting from bevacizumab treatment. Moreover, we highlight current knowledge of the pharmacogenetics of bevacizumab-induced hypertension, which may be used to develop strategies to prevent or minimize this toxicity.
Collapse
Affiliation(s)
- Megan Li
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Deanna L Kroetz
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, United States.
| |
Collapse
|
153
|
Enseleit F, Michels S, Sudano I, Stahel M, Zweifel S, Schlager O, Becker M, Winnik S, Nägele M, Flammer AJ, Neidhart M, Graf N, Matter CM, Seifert B, Lüscher TF, Ruschitzka F. SAVE-AMD: Safety of VEGF Inhibitors in Age-Related Macular Degeneration. Ophthalmologica 2017; 238:205-216. [PMID: 28866675 DOI: 10.1159/000478665] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 06/12/2017] [Indexed: 11/19/2022]
Abstract
OBJECTIVE To determine whether intraocular treatment with vascular endothelial growth factor (VEGF) inhibitors change systemic endothelial function (EF) in patients with neovascular age-related macular degeneration (AMD). METHODS In this prospective, randomized, 2-center, double-masked controlled interventional trial, patients with neovascular and dry AMD were enrolled. Eligible neovascular AMD patients received 2 intravitreal loading doses of either ranibizumab 0.5 mg or bevacizumab 1.25 mg at 4-week intervals and were subsequently followed every 4 weeks and treated according to a pro re nata regime for up to 1 year. Patients with dry AMD served as controls. The primary endpoint was the change in EF assessed by flow-mediated dilatation (FMD) after 2 months of treatment with VEGF inhibitors in patients with AMD compared to patients with dry AMD. FMD was assessed with B-mode high-resolution ultrasonography of the left brachial artery. RESULTS 24 patients with neovascular AMD and 26 patients with dry ADM were included in the trial. Treatment with VEGF inhibitors did not significantly change FMD (from 4.7 ± 2.4 to 3.9 ± 1.9% after 8 weeks, p = 0.07, and to 5.1 ± 2.0% after 1 year; p = 0.93 vs. baseline, respectively). CONCLUSIONS EF did not significantly differ between patients with neovascular AMD treated with intravitreal VEGF inhibition and patients with dry AMD.
Collapse
Affiliation(s)
- Frank Enseleit
- Department of Ophthalmology, City Hospital Triemli Zurich, Zurich, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
154
|
Bernier-Latmani J, Petrova TV. Intestinal lymphatic vasculature: structure, mechanisms and functions. Nat Rev Gastroenterol Hepatol 2017; 14:510-526. [PMID: 28655884 DOI: 10.1038/nrgastro.2017.79] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The mammalian intestine is richly supplied with lymphatic vasculature, which has functions ranging from maintenance of interstitial fluid balance to transport of antigens, antigen-presenting cells, dietary lipids and fat-soluble vitamins. In this Review, we provide in-depth information concerning the organization and structure of intestinal lymphatics, the current view of their developmental origins, as well as molecular mechanisms of intestinal lymphatic patterning and maintenance. We will also discuss physiological aspects of intestinal lymph flow regulation and the known and emerging roles of intestinal lymphatic vessels in human diseases, such as IBD, infection and cancer.
Collapse
Affiliation(s)
- Jeremiah Bernier-Latmani
- Department of Fundamental Oncology, Ludwig Institute for Cancer Research and Institute of Pathology, Centre Hospitalier Universitaire Vaudois and University of Lausanne (UNIL), Chemin des Boveresses 155, Epalinges, Switzerland
| | - Tatiana V Petrova
- Department of Fundamental Oncology, Ludwig Institute for Cancer Research and Institute of Pathology, Centre Hospitalier Universitaire Vaudois and University of Lausanne (UNIL), Chemin des Boveresses 155, Epalinges, Switzerland.,Swiss Institute for Experimental Cancer Research, School of Life Sciences, Swiss Federal Institute of Technology Lausanne, Route Cantonale 1015, Lausanne, Switzerland
| |
Collapse
|
155
|
Clinical pharmacology of anti-angiogenic drugs in oncology. Crit Rev Oncol Hematol 2017; 119:75-93. [PMID: 28916378 DOI: 10.1016/j.critrevonc.2017.08.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 08/23/2017] [Accepted: 08/29/2017] [Indexed: 12/14/2022] Open
Abstract
Abnormal vasculature proliferation is one of the so-called hallmarks of cancer. Angiogenesis inhibitor therapies are one of the major breakthroughs in cancer treatment in the last two decades. Two types of anti-angiogenics have been approved: monoclonal antibodies and derivatives, which are injected and target the extracellular part of a receptor, and protein kinase inhibitors, which are orally taken small molecules targeting the intra-cellular Adenosine Triphosphate -pocket of different kinases. They have become an important part of some tumors' treatment, both in monotherapy or in combination. In this review, we discuss the key pharmacological concepts and the major pitfalls of anti-angiogenic prescriptions. We also review the pharmacokinetic and pharmacodynamics profile of all approved anti-angiogenic protein kinase inhibitors and the potential role of surrogate markers and of therapeutic drug monitoring.
Collapse
|
156
|
Schlecht A, Leimbeck SV, Jägle H, Feuchtinger A, Tamm ER, Braunger BM. Deletion of Endothelial Transforming Growth Factor-β Signaling Leads to Choroidal Neovascularization. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2570-2589. [PMID: 28823871 DOI: 10.1016/j.ajpath.2017.06.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 06/12/2017] [Accepted: 06/29/2017] [Indexed: 12/13/2022]
Abstract
The molecular pathogenesis of choroidal neovascularization (CNV), an angiogenic process that critically contributes to vision loss in age-related macular degeneration, is unclear. Herein, we analyzed the role of transforming growth factor (TGF)-β signaling for CNV formation by generating a series of mutant mouse models with induced conditional deletion of TGF-β signaling in the entire eye, the retinal pigment epithelium (RPE), or the vascular endothelium. Deletion of TGF-β signaling in the eye caused CNV, irrespectively if it was ablated in newborn or 3-week-old mice. Areas of CNV showed photoreceptor degeneration, multilayered RPE, basal lamina deposits, and accumulations of monocytes/macrophages. The changes progressed, leading to marked structural and functional alterations of the retina. Although the specific deletion of TGF-β signaling in the RPE caused no obvious changes, specific deletion in vascular endothelial cells caused CNV and a phenotype similar to that observed after the deletion in the entire eye. We conclude that impairment of TGF-β signaling in the vascular endothelium of the eye is sufficient to trigger CNV formation. Our findings highlight the importance of TGF-β signaling as a key player in the development of ocular neovascularization and indicate a fundamental role of TGF-β signaling in the pathogenesis of age-related macular degeneration.
Collapse
Affiliation(s)
- Anja Schlecht
- Institute of Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany
| | - Sarah V Leimbeck
- Institute of Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany
| | - Herbert Jägle
- Department of Ophthalmology, University Clinic Regensburg, Regensburg, Germany
| | - Annette Feuchtinger
- Research Unit Analytical Pathology, Helmholtz Zentrum Munich, Munich, Germany
| | - Ernst R Tamm
- Institute of Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany.
| | - Barbara M Braunger
- Institute of Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany.
| |
Collapse
|
157
|
Chang ST, Menias CO, Lubner MG, Mellnick VM, Hara AK, Desser TS. Molecular and Clinical Approach to Intra-abdominal Adverse Effects of Targeted Cancer Therapies. Radiographics 2017; 37:1461-1482. [PMID: 28753381 DOI: 10.1148/rg.2017160162] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Targeted cancer therapies encompass an exponentially growing number of agents that involve a myriad of molecular pathways. To excel within this rapidly changing field of clinical oncology, radiologists must eschew traditional organ system-based approaches of cataloging adverse effects in favor of a conceptual framework that incorporates molecular mechanisms and associated clinical outcomes. Understanding molecular mechanisms that underlie imaging manifestations of adverse effects and known associations with treatment response allows radiologists to more effectively recognize adverse effects and differentiate them from tumor progression. Radiologists can therefore more effectively guide oncologists in the management of adverse effects and treatment decisions regarding continuation or cessation of drug therapy. Adverse effects from targeted cancer therapies can be classified into four categories: (a) category 1, on-target adverse effects associated with treatment response; (b) category 2, on-target adverse effects without associated treatment response; (c) category 3, off-target adverse effects; and (d) category 4, tumor necrosis-related adverse effects. This review focuses on adverse effects primarily within the abdomen and pelvis classified according to established or hypothesized molecular mechanisms and illustrated with images of classic examples and several potential emerging toxic effects. ©RSNA, 2017.
Collapse
Affiliation(s)
- Stephanie T Chang
- From the Department of Radiology, VA Palo Alto Health Care System, Palo Alto, Calif (S.T.C.); Department of Radiology, Stanford University School of Medicine, 300 Pasteur Dr, H1307 MC 5621, Stanford, CA 94305 (S.T.C., T.S.D.); Department of Radiology, Mayo Clinic, Scottsdale, Ariz (C.O.M., A.K.H.); Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wis (M.G.L.); and Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Mo (V.M.M.)
| | - Christine O Menias
- From the Department of Radiology, VA Palo Alto Health Care System, Palo Alto, Calif (S.T.C.); Department of Radiology, Stanford University School of Medicine, 300 Pasteur Dr, H1307 MC 5621, Stanford, CA 94305 (S.T.C., T.S.D.); Department of Radiology, Mayo Clinic, Scottsdale, Ariz (C.O.M., A.K.H.); Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wis (M.G.L.); and Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Mo (V.M.M.)
| | - Meghan G Lubner
- From the Department of Radiology, VA Palo Alto Health Care System, Palo Alto, Calif (S.T.C.); Department of Radiology, Stanford University School of Medicine, 300 Pasteur Dr, H1307 MC 5621, Stanford, CA 94305 (S.T.C., T.S.D.); Department of Radiology, Mayo Clinic, Scottsdale, Ariz (C.O.M., A.K.H.); Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wis (M.G.L.); and Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Mo (V.M.M.)
| | - Vincent M Mellnick
- From the Department of Radiology, VA Palo Alto Health Care System, Palo Alto, Calif (S.T.C.); Department of Radiology, Stanford University School of Medicine, 300 Pasteur Dr, H1307 MC 5621, Stanford, CA 94305 (S.T.C., T.S.D.); Department of Radiology, Mayo Clinic, Scottsdale, Ariz (C.O.M., A.K.H.); Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wis (M.G.L.); and Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Mo (V.M.M.)
| | - Amy K Hara
- From the Department of Radiology, VA Palo Alto Health Care System, Palo Alto, Calif (S.T.C.); Department of Radiology, Stanford University School of Medicine, 300 Pasteur Dr, H1307 MC 5621, Stanford, CA 94305 (S.T.C., T.S.D.); Department of Radiology, Mayo Clinic, Scottsdale, Ariz (C.O.M., A.K.H.); Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wis (M.G.L.); and Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Mo (V.M.M.)
| | - Terry S Desser
- From the Department of Radiology, VA Palo Alto Health Care System, Palo Alto, Calif (S.T.C.); Department of Radiology, Stanford University School of Medicine, 300 Pasteur Dr, H1307 MC 5621, Stanford, CA 94305 (S.T.C., T.S.D.); Department of Radiology, Mayo Clinic, Scottsdale, Ariz (C.O.M., A.K.H.); Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wis (M.G.L.); and Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Mo (V.M.M.)
| |
Collapse
|
158
|
Ayar O, Alpay A, Koban Y, Akdemir MO, Yazgan S, Canturk Ugurbas S, Ugurbas SH. The Effect of Dexamethasone Intravitreal Implant on Retinal Nerve Fiber Layer in Patients Diagnosed with Branch Retinal Vein Occlusion. Curr Eye Res 2017. [PMID: 28632411 DOI: 10.1080/02713683.2017.1313430] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
PURPOSE To evaluate the effect of a single dose of intravitreal dexamethasone (DEX) implant on retinal nerve fiber layer (RNFL) thickness in patients with branch retinal vein occlusion (BRVO) in a 6-month period. MATERIALS AND METHODS This retrospective observational study included the patients with BRVO who received intravitreal DEX implant and whose assessment included the baseline RNFL thickness measurements. The data of 26 eyes of 24 patients were retrospectively analyzed. Spectral domain optic coherence tomography was used to measure peripapillary RNFL thickness in six regional subfields. Intraocular pressure (IOP) values at each visit were recorded. The data of single dose DEX implant during 6 months were assessed. RESULTS The mean preoperative and postoperative 6th month nasal RNFL values were 85.4 ± 23.0 μm and 82.1 ± 17.6 μm, respectively, and the difference between the measurements was not statistically significant (p = 0.372). There was a slight decrease in the mean RNFL values postoperatively compared to the baseline values in all quadrants except supero-temporal quadrant; however, none of them reached statistically significant level (p > 0.05). The mean IOP values before and 6 months after implantation were 15.7 ± 2.9 mmHg and 16.5 ± 4.2 mmHg, respectively. The difference between the 6th month IOP values and baseline IOP values was not statistically significant (p = 0.236). CONCLUSION Intravitreal DEX implant seems to have no adverse effect on RNFL thickness in BRVO patients in a 6-month period.
Collapse
Affiliation(s)
- Orhan Ayar
- a Faculty of Medicine, Department of Ophthalmology , Bulent Ecevit University , Zonguldak , Turkey
| | - Atilla Alpay
- a Faculty of Medicine, Department of Ophthalmology , Bulent Ecevit University , Zonguldak , Turkey
| | - Yaran Koban
- b Faculty of Medicine, Department of Ophthalmology , Kafkas University , Kars , Turkey
| | - Mehmet Orcun Akdemir
- a Faculty of Medicine, Department of Ophthalmology , Bulent Ecevit University , Zonguldak , Turkey
| | - Serpil Yazgan
- a Faculty of Medicine, Department of Ophthalmology , Bulent Ecevit University , Zonguldak , Turkey
| | - Sılay Canturk Ugurbas
- a Faculty of Medicine, Department of Ophthalmology , Bulent Ecevit University , Zonguldak , Turkey
| | - Suat Hayri Ugurbas
- a Faculty of Medicine, Department of Ophthalmology , Bulent Ecevit University , Zonguldak , Turkey
| |
Collapse
|
159
|
Maintenance of antiangiogenic and antitumor effects by orally active low-dose capecitabine for long-term cancer therapy. Proc Natl Acad Sci U S A 2017; 114:E5226-E5235. [PMID: 28607065 DOI: 10.1073/pnas.1705066114] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Long-term uninterrupted therapy is essential for maximizing clinical benefits of antiangiogenic drugs (AADs) in cancer patients. Unfortunately, nearly all clinically available AADs are delivered to cancer patients using disrupted regimens. We aim to develop lifetime, nontoxic, effective, orally active, and low-cost antiangiogenic and antitumor drugs for treatment of cancer patients. Here we report our findings of long-term maintenance therapy with orally active, nontoxic, low cost antiangiogenic chemotherapeutics for effective cancer treatment. In a sequential treatment regimen, robust antiangiogenic effects in tumors were achieved with an anti-VEGF drug, followed by a low-dose chemotherapy. The nontoxic, low dose of the orally active prodrug capecitabine was able to sustain the anti-VEGF-induced vessel regression for long periods. In another experimental setting, maintenance of low-dose capecitabine produced greater antiangiogenic and antitumor effects after AAD plus chemotherapy. No obvious adverse effects were developed after more than 2-mo of consecutive treatment with a low dose of capecitabine. Together, our findings provide a rationalized concept of effective cancer therapy by long-term maintenance of AAD-triggered antiangiogenic effects with orally active, nontoxic, low-cost, clinically available chemotherapeutics.
Collapse
|
160
|
Matrone A, Valerio L, Pieruzzi L, Giani C, Cappagli V, Lorusso L, Agate L, Puleo L, Viola D, Bottici V, Del Re M, Molinaro E, Danesi R, Elisei R. Protein kinase inhibitors for the treatment of advanced and progressive radiorefractory thyroid tumors: From the clinical trials to the real life. Best Pract Res Clin Endocrinol Metab 2017; 31:319-334. [PMID: 28911728 DOI: 10.1016/j.beem.2017.06.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The last ten years have been characterized by the introduction in the clinical practice of new drugs named tyrosine kinase inhibitors for the treatment of several human tumors. After the positive conclusion of two international multicentric, randomized phase III clinical trials, two of these drugs, sorafenib and lenvatinib, have been recently approved and they are now available for the treatment of advanced and progressive radioiodine refractory thyroid tumors. We have been involved in most clinical trials performed with different tyrosine kinase inhibitors in different histotypes of thyroid cancer thus acquiring a lot of experience in the management of both drugs and their adverse events. Aim of this review is to give an overview of both the rationale for the use of these inhibitors in thyroid cancer and the major results of the clinical trials. Some suggestions for the management of treated patients in the real life are also provided.
Collapse
Affiliation(s)
- Antonio Matrone
- Unit of Endocrinology, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Laura Valerio
- Unit of Endocrinology, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Letizia Pieruzzi
- Unit of Endocrinology, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Carlotta Giani
- Unit of Endocrinology, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Virginia Cappagli
- Unit of Endocrinology, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Loredana Lorusso
- Unit of Endocrinology, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Laura Agate
- Unit of Endocrinology, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Luciana Puleo
- Unit of Endocrinology, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - David Viola
- Unit of Endocrinology, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Valeria Bottici
- Unit of Endocrinology, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Marzia Del Re
- Unit of Pharmacology, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Eleonora Molinaro
- Unit of Endocrinology, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Romano Danesi
- Unit of Pharmacology, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Rossella Elisei
- Unit of Endocrinology, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Via Paradisa 2, 56124 Pisa, Italy.
| |
Collapse
|
161
|
Vascular heterogeneity and specialization in development and disease. Nat Rev Mol Cell Biol 2017; 18:477-494. [PMID: 28537573 DOI: 10.1038/nrm.2017.36] [Citation(s) in RCA: 394] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Blood and lymphatic vessels pervade almost all body tissues and have numerous essential roles in physiology and disease. The inner lining of these networks is formed by a single layer of endothelial cells, which is specialized according to the needs of the tissue that it supplies. Whereas the general mechanisms of blood and lymphatic vessel development are being defined with increasing molecular precision, studies of the processes of endothelial specialization remain mostly descriptive. Recent insights from genetic animal models illuminate how endothelial cells interact with each other and with their tissue environment, providing paradigms for vessel type- and organ-specific endothelial differentiation. Delineating these governing principles will be crucial for understanding how tissues develop and maintain, and how their function becomes abnormal in disease.
Collapse
|
162
|
Saharinen P, Eklund L, Alitalo K. Therapeutic targeting of the angiopoietin-TIE pathway. Nat Rev Drug Discov 2017; 16:635-661. [PMID: 28529319 DOI: 10.1038/nrd.2016.278] [Citation(s) in RCA: 375] [Impact Index Per Article: 46.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The endothelial angiopoietin (ANG)-TIE growth factor receptor pathway regulates vascular permeability and pathological vascular remodelling during inflammation, tumour angiogenesis and metastasis. Drugs that target the ANG-TIE pathway are in clinical development for oncological and ophthalmological applications. The aim is to complement current vascular endothelial growth factor (VEGF)-based anti-angiogenic therapies in cancer, wet age-related macular degeneration and macular oedema. The unique function of the ANG-TIE pathway in vascular stabilization also renders this pathway an attractive target in sepsis, organ transplantation, atherosclerosis and vascular complications of diabetes. This Review covers key aspects of the function of the ANG-TIE pathway in vascular disease and describes the recent development of novel therapeutics that target this pathway.
Collapse
Affiliation(s)
- Pipsa Saharinen
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, Haartmaninkatu 8, P.O. Box 63, FI-00014 Helsinki, Finland
| | - Lauri Eklund
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, Aapistie 5A, University of Oulu, 90220 Oulu, Finland
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, Haartmaninkatu 8, P.O. Box 63, FI-00014 Helsinki, Finland
| |
Collapse
|
163
|
Castro J, Puente P, Martínez R, Hernández A, Morera Y, Martínez L, Aldana L, Valdés I, Ayala M, Cosme K. Vaccine CIGB 247 is potentially safe for use as a novel therapeutic vaccine against cancer in Chlorocebus aethiops monkeys. Int Immunopharmacol 2017; 48:55-60. [PMID: 28463787 DOI: 10.1016/j.intimp.2017.04.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 03/31/2017] [Accepted: 04/22/2017] [Indexed: 01/22/2023]
Abstract
CIGB 247 is a novel cancer therapeutic vaccine based on human vascular endothelial growth factor (VEGF) variant molecule as antigen, in combination with a bacterial adjuvant. This vaccine candidate has previously demonstrated efficacy and safety in mice, rats, rabbits and non-human primates. In the present study we evaluated the effects on the clinical, hematological and biochemical parameters of CIGB 247 vaccine in Chlorocebus aethiops monkeys. Three groups of monkeys were immunized with three doses of vaccine formulation to measure physiological values of clinical, hematological and serum biochemical parameters. Monkeys' body weight and temperature were kept stable and close to standard values throughout the study. Variations in the levels of red blood cells and hemoglobin were observed among the different groups for all injected doses, but these hematological parameters recovered normal values at the end of the study. On the other hand, biochemical parameters such as the total bilirubin and total protein counts showed variations along the study, while they were not associated with the test substance. In summary, no negative effects on clinical, hematological and biochemical parameters were detected. Together, our results put forward the potential and support the safety of the CIGB 247 vaccine candidate for use in clinical applications. The data presented here can be used to estimate a human dosing regimen from preclinical data.
Collapse
Affiliation(s)
- Jorge Castro
- Center for Genetic Engineering and Biotechnology (CIGB), Avenue 31, PO. Box 6162, Havana 6 10600, Cuba.
| | - Pedro Puente
- Center for Genetic Engineering and Biotechnology (CIGB), Avenue 31, PO. Box 6162, Havana 6 10600, Cuba
| | - Rafael Martínez
- Center for Genetic Engineering and Biotechnology (CIGB), Avenue 31, PO. Box 6162, Havana 6 10600, Cuba
| | - Alexander Hernández
- Center for Genetic Engineering and Biotechnology (CIGB), Avenue 31, PO. Box 6162, Havana 6 10600, Cuba
| | - Yanelys Morera
- Center for Genetic Engineering and Biotechnology (CIGB), Avenue 31, PO. Box 6162, Havana 6 10600, Cuba
| | - Leticia Martínez
- Center for Genetic Engineering and Biotechnology (CIGB), Avenue 31, PO. Box 6162, Havana 6 10600, Cuba
| | - Lizet Aldana
- Center for Genetic Engineering and Biotechnology (CIGB), Avenue 31, PO. Box 6162, Havana 6 10600, Cuba
| | - Iris Valdés
- Center for Genetic Engineering and Biotechnology (CIGB), Avenue 31, PO. Box 6162, Havana 6 10600, Cuba
| | - Marta Ayala
- Center for Genetic Engineering and Biotechnology (CIGB), Avenue 31, PO. Box 6162, Havana 6 10600, Cuba
| | - Karelia Cosme
- Center for Genetic Engineering and Biotechnology (CIGB), Avenue 31, PO. Box 6162, Havana 6 10600, Cuba
| |
Collapse
|
164
|
Rojas-Canales D, Penko D, Myo Min KK, Parham KA, Peiris H, Haberberger RV, Pitson SM, Drogemuller C, Keating DJ, Grey ST, Coates PT, Bonder CS, Jessup CF. Local Sphingosine Kinase 1 Activity Improves Islet Transplantation. Diabetes 2017; 66:1301-1311. [PMID: 28174291 DOI: 10.2337/db16-0837] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Accepted: 02/02/2017] [Indexed: 11/13/2022]
Abstract
Pancreatic islet transplantation is a promising clinical treatment for type 1 diabetes, but success is limited by extensive β-cell death in the immediate posttransplant period and impaired islet function in the longer term. Following transplantation, appropriate vascular remodeling is crucial to ensure the survival and function of engrafted islets. The sphingosine kinase (SK) pathway is an important regulator of vascular beds, but its role in the survival and function of transplanted islets is unknown. We observed that donor islets from mice deficient in SK1 (Sphk1 knockout) contain a reduced number of resident intraislet vascular endothelial cells. Furthermore, we demonstrate that the main product of SK1, sphingosine-1-phosphate, controls the migration of intraislet endothelial cells in vitro. We reveal in vivo that Sphk1 knockout islets have an impaired ability to cure diabetes compared with wild-type controls. Thus, SK1-deficient islets not only contain fewer resident vascular cells that participate in revascularization, but likely also a reduced ability to recruit new vessels into the transplanted islet. Together, our data suggest that SK1 is important for islet revascularization following transplantation and represents a novel clinical target for improving transplant outcomes.
Collapse
Affiliation(s)
- Darling Rojas-Canales
- Discipline of Medicine, The University of Adelaide, Adelaide, Australia
- Central Northern Adelaide Renal and Transplantation Services, Royal Adelaide Hospital, Adelaide, Australia
| | - Daniella Penko
- Discipline of Medicine, The University of Adelaide, Adelaide, Australia
- Central Northern Adelaide Renal and Transplantation Services, Royal Adelaide Hospital, Adelaide, Australia
| | - Kay K Myo Min
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia
| | - Kate A Parham
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia
| | - Heshan Peiris
- Department of Human Physiology, Flinders University, Bedford Park, Australia
- Centre for Neuroscience, Flinders University, Bedford Park, Australia
| | | | - Stuart M Pitson
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia
| | - Chris Drogemuller
- Discipline of Medicine, The University of Adelaide, Adelaide, Australia
- Central Northern Adelaide Renal and Transplantation Services, Royal Adelaide Hospital, Adelaide, Australia
| | - Damien J Keating
- Department of Human Physiology, Flinders University, Bedford Park, Australia
- Centre for Neuroscience, Flinders University, Bedford Park, Australia
- South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Shane T Grey
- Garvan Medical Institute, Darlinghurst, Australia
| | - Patrick T Coates
- Discipline of Medicine, The University of Adelaide, Adelaide, Australia
- Central Northern Adelaide Renal and Transplantation Services, Royal Adelaide Hospital, Adelaide, Australia
| | - Claudine S Bonder
- Discipline of Medicine, The University of Adelaide, Adelaide, Australia
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia
| | - Claire F Jessup
- Discipline of Medicine, The University of Adelaide, Adelaide, Australia
- Centre for Neuroscience, Flinders University, Bedford Park, Australia
- Department of Anatomy & Histology, Flinders University, Bedford Park, Australia
| |
Collapse
|
165
|
Lutz SZ, Ullrich A, Häring HU, Ullrich S, Gerst F. Sunitinib specifically augments glucose-induced insulin secretion. Cell Signal 2017; 36:91-97. [PMID: 28449948 DOI: 10.1016/j.cellsig.2017.04.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 04/07/2017] [Accepted: 04/23/2017] [Indexed: 01/01/2023]
Abstract
The tyrosine kinase inhibitor sunitinib is used for the treatment of numerous cancers in humans. In diabetic patients, sunitinib lowers blood glucose levels and improves glycaemic control. This study aims to analyse whether sunitinib has specific and direct effects on insulin secreting β-cells. Regulation of insulin secretion, of cellular cAMP levels and activation of signalling pathways were examined upon exposure of rat insulinoma INS-1E cells to sunitinib under specific stimulatory and inhibitory conditions. Secreted insulin and cellular cAMP levels were measured using RIA and ELISA, respectively. Protein phosphorylations were examined on western blots. Sunitinib enhanced glucose-induced insulin secretion (GIIS) concentration-dependently, reaching a maximal stimulation at 2μM. Sunitinib further augmented insulin secretion in the presence of elevated cAMP levels and the FFAR1 agonists. Adrenaline and the PKA inhibitor H89 counteracted the stimulatory effect of sunitinib on secretion. However, sunitinib altered neither the cellular levels of cAMP nor the phosphorylation of PKA. Sunitinib did not reduce IGF-1-induced phosphorylation of AKT/PKB and ERK1/2. In conclusion, these results suggest that sunitinib stimulates GIIS by a direct effect on β-cells, which may contribute to the glucose-lowering action of the tyrosine kinase inhibitor in humans.
Collapse
Affiliation(s)
- Stefan Z Lutz
- German Center for Diabetes Research (DZD e.V.), Germany; Institute for Diabetes Research and Metabolic Diseases IDM of the Helmholtz Center Munich at the Eberhard-Karls-University of Tübingen, Germany; University Hospital Tübingen, Internal Medicine IV, Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Otfried-Müller-Str. 10, 72076 Tübingen, Germany
| | - Axel Ullrich
- Department of Molecular Biology, Max-Planck-Institute of Biochemistry, Martinsried, Germany
| | - Hans-Ulrich Häring
- German Center for Diabetes Research (DZD e.V.), Germany; Institute for Diabetes Research and Metabolic Diseases IDM of the Helmholtz Center Munich at the Eberhard-Karls-University of Tübingen, Germany; University Hospital Tübingen, Internal Medicine IV, Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Otfried-Müller-Str. 10, 72076 Tübingen, Germany
| | - Susanne Ullrich
- German Center for Diabetes Research (DZD e.V.), Germany; Institute for Diabetes Research and Metabolic Diseases IDM of the Helmholtz Center Munich at the Eberhard-Karls-University of Tübingen, Germany; University Hospital Tübingen, Internal Medicine IV, Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Otfried-Müller-Str. 10, 72076 Tübingen, Germany
| | - Felicia Gerst
- German Center for Diabetes Research (DZD e.V.), Germany; Institute for Diabetes Research and Metabolic Diseases IDM of the Helmholtz Center Munich at the Eberhard-Karls-University of Tübingen, Germany; University Hospital Tübingen, Internal Medicine IV, Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Otfried-Müller-Str. 10, 72076 Tübingen, Germany.
| |
Collapse
|
166
|
Narayanan S, Loganathan G, Dhanasekaran M, Tucker W, Patel A, Subhashree V, Mokshagundam S, Hughes MG, Williams SK, Balamurugan AN. Intra-islet endothelial cell and β-cell crosstalk: Implication for islet cell transplantation. World J Transplant 2017; 7:117-128. [PMID: 28507914 PMCID: PMC5409911 DOI: 10.5500/wjt.v7.i2.117] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 02/28/2017] [Accepted: 03/24/2017] [Indexed: 02/05/2023] Open
Abstract
The intra-islet microvasculature is a critical interface between the blood and islet endocrine cells governing a number of cellular and pathophysiological processes associated with the pancreatic tissue. A growing body of evidence indicates a strong functional and physical interdependency of β-cells with endothelial cells (ECs), the building blocks of islet microvasculature. Intra-islet ECs, actively regulate vascular permeability and appear to play a role in fine-tuning blood glucose sensing and regulation. These cells also tend to behave as “guardians”, controlling the expression and movement of a number of important immune mediators, thereby strongly contributing to the physiology of islets. This review will focus on the molecular signalling and crosstalk between the intra-islet ECs and β-cells and how their relationship can be a potential target for intervention strategies in islet pathology and islet transplantation.
Collapse
|
167
|
Horizontal alignment of 5' -> 3' intergene distance segment tropy with respect to the gene as the conserved basis for DNA transcription. Future Sci OA 2017; 3:FSO160. [PMID: 28344824 PMCID: PMC5351715 DOI: 10.4155/fsoa-2016-0070] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 10/31/2016] [Indexed: 01/31/2023] Open
Abstract
AIM To study the conserved basis for gene expression in comparative cell types at opposite ends of the cell pressuromodulation spectrum, the lymphatic endothelial cell and the blood microvascular capillary endothelial cell. METHODS The mechanism for gene expression is studied in terms of the 5' -> 3' direction paired point tropy quotients (prpTQs) and the final 5' -> 3' direction episodic sub-episode block sums split-integrated weighted average-averaged gene overexpression tropy quotient (esebssiwaagoTQ). RESULTS The final 5' -> 3' esebssiwaagoTQ classifies an lymphatic endothelial cell overexpressed gene as a supra-pressuromodulated gene (esebssiwaagoTQ ≥ 0.25 < 0.75) every time and classifies a blood microvascular capillary endothelial cell overexpressed gene every time as an infra-pressuromodulated gene (esebssiwaagoTQ < 0.25) (100% sensitivity; 100% specificity). CONCLUSION Horizontal alignment of 5' -> 3' intergene distance segment tropy wrt the gene is the basis for DNA transcription in the pressuromodulated state.
Collapse
|
168
|
Chauvet N, Romanò N, Lafont C, Guillou A, Galibert E, Bonnefont X, Le Tissier P, Fedele M, Fusco A, Mollard P, Coutry N. Complementary actions of dopamine D2 receptor agonist and anti-vegf therapy on tumoral vessel normalization in a transgenic mouse model. Int J Cancer 2017; 140:2150-2161. [PMID: 28152577 DOI: 10.1002/ijc.30628] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 01/11/2017] [Accepted: 01/25/2017] [Indexed: 12/21/2022]
Abstract
Angiogenesis contributes in multiple ways to disease progression in tumors and reduces treatment efficiency. Molecular therapies targeting Vegf signaling combined with chemotherapy or other drugs exhibit promising results to improve efficacy of treatment. Dopamine has been recently proposed to be a novel safe anti-angiogenic drug that stabilizes abnormal blood vessels and increases therapeutic efficacy. Here, we aimed to identify a treatment to normalize tumoral vessels and restore normal blood perfusion in tumor tissue with a Vegf receptor inhibitor and/or a ligand of dopamine G protein-coupled receptor D2 (D2R). Dopamine, via its action on D2R, is an endogenous effector of the pituitary gland, and we took advantage of this system to address this question. We have used a previously described Hmga2/T mouse model developing haemorrhagic prolactin-secreting adenomas. In mutant mice, blood vessels are profoundly altered in tumors, and an aberrant arterial vascularization develops leading to the loss of dopamine supply. D2R agonist treatment blocks tumor growth, induces regression of the aberrant blood supply and normalizes blood vessels. A chronic treatment is able to restore the altered balance between pro- and anti-angiogenic factors. Remarkably, an acute treatment induces an upregulation of the stabilizing factor Angiopoietin 1. An anti-Vegf therapy is also effective to restrain tumor growth and improves vascular remodeling. Importantly, only the combination treatment suppresses intratumoral hemorrhage and restores blood vessel perfusion, suggesting that it might represent an attractive therapy targeting tumor vasculature. Similar strategies targeting other ligands of GPCRs involved in angiogenesis may identify novel therapeutic opportunities for cancer.
Collapse
Affiliation(s)
- Norbert Chauvet
- CNRS, UMR-5203, Institut de Génomique Fonctionnelle, Département de Physiologie, Montpellier, F-34094, France.,INSERM, U1191, Montpellier, F-34094, France.,Université de Montpellier, UMR-5203, Montpellier, F-34094, France
| | - Nicola Romanò
- CNRS, UMR-5203, Institut de Génomique Fonctionnelle, Département de Physiologie, Montpellier, F-34094, France.,INSERM, U1191, Montpellier, F-34094, France.,Université de Montpellier, UMR-5203, Montpellier, F-34094, France
| | - Chrystel Lafont
- CNRS, UMR-5203, Institut de Génomique Fonctionnelle, Département de Physiologie, Montpellier, F-34094, France.,INSERM, U1191, Montpellier, F-34094, France.,Université de Montpellier, UMR-5203, Montpellier, F-34094, France
| | - Anne Guillou
- CNRS, UMR-5203, Institut de Génomique Fonctionnelle, Département de Physiologie, Montpellier, F-34094, France.,INSERM, U1191, Montpellier, F-34094, France.,Université de Montpellier, UMR-5203, Montpellier, F-34094, France
| | - Evelyne Galibert
- CNRS, UMR-5203, Institut de Génomique Fonctionnelle, Département de Physiologie, Montpellier, F-34094, France.,INSERM, U1191, Montpellier, F-34094, France.,Université de Montpellier, UMR-5203, Montpellier, F-34094, France
| | - Xavier Bonnefont
- CNRS, UMR-5203, Institut de Génomique Fonctionnelle, Département de Physiologie, Montpellier, F-34094, France.,INSERM, U1191, Montpellier, F-34094, France.,Université de Montpellier, UMR-5203, Montpellier, F-34094, France
| | - Paul Le Tissier
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, EH8 9XD, United Kingdom
| | - Monica Fedele
- Istituto di Endocrinologia ed Oncologia Sperimentale del CNR e/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", Naples, 80131, Italy
| | - Alfredo Fusco
- Istituto di Endocrinologia ed Oncologia Sperimentale del CNR e/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", Naples, 80131, Italy
| | - Patrice Mollard
- CNRS, UMR-5203, Institut de Génomique Fonctionnelle, Département de Physiologie, Montpellier, F-34094, France.,INSERM, U1191, Montpellier, F-34094, France.,Université de Montpellier, UMR-5203, Montpellier, F-34094, France
| | - Nathalie Coutry
- CNRS, UMR-5203, Institut de Génomique Fonctionnelle, Département de Physiologie, Montpellier, F-34094, France.,INSERM, U1191, Montpellier, F-34094, France.,Université de Montpellier, UMR-5203, Montpellier, F-34094, France
| |
Collapse
|
169
|
Hargreaves A, Bigley A, Price S, Kendrew J, Barry ST. Automated image analysis of intra-tumoral and peripheral endocrine organ vascular bed regression using 'Fibrelength' as a novel structural biomarker. J Appl Toxicol 2017; 37:902-912. [PMID: 28186326 DOI: 10.1002/jat.3438] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 12/15/2016] [Accepted: 12/15/2016] [Indexed: 12/16/2022]
Abstract
The study of vascular modulation has received a great deal of attention in recent years as knowledge has increased around the role of angiogenesis within disease contexts such as cancer. Despite rapidly expanding insights into the molecular processes involved and the concomitant generation of a number of anticancer vascular modulating chemotherapeutics, techniques used in the measurement of structural vascular change have advanced more modestly, particularly with regard to the preclinical quantification of off-target vascular regression within systemic, notably endocrine, blood vessels. Such changes translate into a number of major clinical side effects and there remains a need for improved preclinical screening and analysis. Here we present the generation of a novel structural biomarker, which can be incorporated into a number of contemporary image analysis platforms and used to compare tumour versus systemic host tissue vascularity. By contrasting the measurements obtained, the preclinical efficacy of vascular modulating chemotherapies can be evaluated in light of the predicted therapeutic window. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Adam Hargreaves
- PathCelerate Ltd, The BioHub, Alderley Park, Mereside, Alderley Edge, Cheshire, UK
| | - Alison Bigley
- Oraclebio Ltd, BioCity Scotland, North Lanarkshire, Scotland, UK
| | - Shirley Price
- University of Surrey, 3660 Office of the Vice-Provost, Guildford, Surrey, UK
| | - Jane Kendrew
- AstraZeneca PLC, Oncology iMED, Alderley Edge, Cheshire, UK
| | - Simon T Barry
- AstraZeneca PLC, Oncology iMED, Alderley Edge, Cheshire, UK
| |
Collapse
|
170
|
HO-1 inhibits preadipocyte proliferation and differentiation at the onset of obesity via ROS dependent activation of Akt2. Sci Rep 2017; 7:40881. [PMID: 28102348 PMCID: PMC5244367 DOI: 10.1038/srep40881] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 12/12/2016] [Indexed: 01/15/2023] Open
Abstract
Excessive accumulation of white adipose tissue (WAT) is a hallmark of obesity. The expansion of WAT in obesity involves proliferation and differentiation of adipose precursors, however, the underlying molecular mechanisms remain unclear. Here, we used an unbiased transcriptomics approach to identify the earliest molecular underpinnings occuring in adipose precursors following a brief HFD in mice. Our analysis identifies Heme Oxygenase-1 (HO-1) as strongly and selectively being upregulated in the adipose precursor fraction of WAT, upon high-fat diet (HFD) feeding. Specific deletion of HO-1 in adipose precursors of Hmox1fl/flPdgfraCre mice enhanced HFD-dependent visceral adipose precursor proliferation and differentiation. Mechanistically, HO-1 reduces HFD-induced AKT2 phosphorylation via ROS thresholding in mitochondria to reduce visceral adipose precursor proliferation. HO-1 influences adipogenesis in a cell-autonomous way by regulating
events early in adipogenesis, during the process of mitotic clonal expansion, upstream of Cebpα and PPARγ. Similar effects on human preadipocyte proliferation and differentiation in vitro were observed upon modulation of HO-1 expression. This collectively renders HO-1 as an essential factor linking extrinsic factors (HFD) with inhibition of specific downstream molecular mediators (ROS & AKT2), resulting in diminished adipogenesis that may contribute to hyperplastic adipose tissue expansion.
Collapse
|
171
|
Payne SL, Peacock HM, Vickaryous MK. Blood vessel formation during tail regeneration in the leopard gecko (Eublepharis macularius): The blastema is not avascular. J Morphol 2017; 278:380-389. [PMID: 28078708 DOI: 10.1002/jmor.20648] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 12/08/2016] [Accepted: 12/09/2016] [Indexed: 01/08/2023]
Abstract
Unique among amniotes, many lizards are able to self-detach (autotomize) their tail and then regenerate a replacement. Tail regeneration involves the formation of a blastema, an accumulation of proliferating cells at the site of autotomy. Over time, cells of the blastema give rise to most of the tissues in the replacement tail. In non-amniotes capable of regenerating (such as urodeles and some teleost fish), the blastema is reported to be essentially avascular until tissue differentiation takes place. For tail regenerating lizards less is known. Here, we investigate neovascularization during tail regeneration in the leopard gecko (Eublepharis macularius). We demonstrate that the gecko tail blastema is not an avascular structure. Beginning with the onset of regenerative outgrowth, structurally mature (mural cell supported) blood vessels are found within the blastema. Although the pattern of blood vessel distribution in the regenerate tail differs from that of the original, a hierarchical network is established, with vessels of varying luminal diameters and wall thicknesses. Using immunostaining, we determine that blastema outgrowth and tissue differentiation is characterized by a dynamic interplay between the pro-angiogenic protein vascular endothelial growth factor (VEGF) and the anti-angiogenic protein thrombospondin-1 (TSP-1). VEGF-expression is initially widespread, but diminishes as tissues differentiate. In contrast, TSP-1 expression is initially restricted but becomes more abundant as VEGF-expression wanes. We predict that variation in the neovascular response observed between different regeneration-competent species likely relates to the volume of the blastema. J. Morphol. 278:380-389, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Samantha L Payne
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Hanna M Peacock
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Matthew K Vickaryous
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
172
|
Kragl M, Schubert R, Karsjens H, Otter S, Bartosinska B, Jeruschke K, Weiss J, Chen C, Alsteens D, Kuss O, Speier S, Eberhard D, Müller DJ, Lammert E. The biomechanical properties of an epithelial tissue determine the location of its vasculature. Nat Commun 2016; 7:13560. [PMID: 27995929 PMCID: PMC5187430 DOI: 10.1038/ncomms13560] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 10/14/2016] [Indexed: 01/06/2023] Open
Abstract
An important question is how growing tissues establish a blood vessel network. Here we study vascular network formation in pancreatic islets, endocrine tissues derived from pancreatic epithelium. We find that depletion of integrin-linked kinase (ILK) in the pancreatic epithelial cells of mice results in glucose intolerance due to a loss of the intra-islet vasculature. In turn, blood vessels accumulate at the islet periphery. Neither alterations in endothelial cell proliferation, apoptosis, morphology, Vegfa expression and VEGF-A secretion nor ‘empty sleeves' of vascular basement membrane are found. Instead, biophysical experiments reveal that the biomechanical properties of pancreatic islet cells, such as their actomyosin-mediated cortex tension and adhesive forces to endothelial cells, are significantly changed. These results suggest that a sorting event is driving the segregation of endothelial and epithelial cells and indicate that the epithelial biomechanical properties determine whether the blood vasculature invades or envelops a growing epithelial tissue. Vasculature is denser in soft than in stiff tissues. Kragl et al. suggest a mechanistic link between biomechanical tissue properties and vascularization by showing that integrin-linked kinase reduces the contractile forces of the cell cortex in endocrine pancreatic cells, facilitating their adhesion to blood vessels and enabling pancreatic islet vascularization.
Collapse
Affiliation(s)
- Martin Kragl
- Institute of Metabolic Physiology, Department of Biology, Heinrich Heine University, D-40225 Düsseldorf, Germany.,German Center for Diabetes Research (DZD e.V.), D-85764 München-Neuherberg, Germany.,Institute for Beta Cell Biology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, D-40225 Düsseldorf, Germany
| | - Rajib Schubert
- Eidgenössische Technische Hochschule Zürich, Department of Biosystems Science and Engineering, CH-4058 Basel, Switzerland
| | - Haiko Karsjens
- Institute of Metabolic Physiology, Department of Biology, Heinrich Heine University, D-40225 Düsseldorf, Germany.,German Center for Diabetes Research (DZD e.V.), D-85764 München-Neuherberg, Germany.,Institute for Beta Cell Biology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, D-40225 Düsseldorf, Germany
| | - Silke Otter
- Institute of Metabolic Physiology, Department of Biology, Heinrich Heine University, D-40225 Düsseldorf, Germany
| | - Barbara Bartosinska
- Institute of Metabolic Physiology, Department of Biology, Heinrich Heine University, D-40225 Düsseldorf, Germany.,German Center for Diabetes Research (DZD e.V.), D-85764 München-Neuherberg, Germany.,Institute for Beta Cell Biology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, D-40225 Düsseldorf, Germany
| | - Kay Jeruschke
- German Center for Diabetes Research (DZD e.V.), D-85764 München-Neuherberg, Germany.,Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, D-40225 Düsseldorf, Germany
| | - Jürgen Weiss
- German Center for Diabetes Research (DZD e.V.), D-85764 München-Neuherberg, Germany.,Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, D-40225 Düsseldorf, Germany
| | - Chunguang Chen
- German Center for Diabetes Research (DZD e.V.), D-85764 München-Neuherberg, Germany.,Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.,DFG-Center for Regenerative Therapies Dresden (CRTD), Faculty of Medicine, Technische Universität Dresden, D-01307 Dresden, Germany
| | - David Alsteens
- Eidgenössische Technische Hochschule Zürich, Department of Biosystems Science and Engineering, CH-4058 Basel, Switzerland
| | - Oliver Kuss
- German Center for Diabetes Research (DZD e.V.), D-85764 München-Neuherberg, Germany.,Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, D-40225 Düsseldorf, Germany
| | - Stephan Speier
- German Center for Diabetes Research (DZD e.V.), D-85764 München-Neuherberg, Germany.,Paul Langerhans Institute Dresden (PLID) of Helmholtz Center Munich at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, D-85764 Neuherberg, Germany.,DFG-Center for Regenerative Therapies Dresden (CRTD), Faculty of Medicine, Technische Universität Dresden, D-01307 Dresden, Germany
| | - Daniel Eberhard
- Institute of Metabolic Physiology, Department of Biology, Heinrich Heine University, D-40225 Düsseldorf, Germany
| | - Daniel J Müller
- Eidgenössische Technische Hochschule Zürich, Department of Biosystems Science and Engineering, CH-4058 Basel, Switzerland
| | - Eckhard Lammert
- Institute of Metabolic Physiology, Department of Biology, Heinrich Heine University, D-40225 Düsseldorf, Germany.,German Center for Diabetes Research (DZD e.V.), D-85764 München-Neuherberg, Germany.,Institute for Beta Cell Biology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, D-40225 Düsseldorf, Germany
| |
Collapse
|
173
|
Marneros AG. Increased VEGF-A promotes multiple distinct aging diseases of the eye through shared pathomechanisms. EMBO Mol Med 2016; 8:208-31. [PMID: 26912740 PMCID: PMC4772957 DOI: 10.15252/emmm.201505613] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
While increased VEGF‐A has been associated with neovascular age‐related macular degeneration (AMD), it is not known whether VEGF‐A may also promote other age‐related eye diseases. Here, we show that an increase in VEGF‐A is sufficient to cause multiple distinct common aging diseases of the eye, including cataracts and both neovascular and non‐exudative AMD‐like pathologies. In the lens, increased VEGF‐A induces age‐related opacifications that are associated with ERK hyperactivation, increased oxidative damage, and higher expression of the NLRP3 inflammasome effector cytokine IL‐1β. Similarly, increased VEGF‐A induces oxidative stress and IL‐1β expression also in the retinal pigment epithelium (RPE). Targeting NLRP3 inflammasome components or Il1r1 strongly inhibited not only VEGF‐A‐induced cataract formation, but also both neovascular and non‐exudative AMD‐like pathologies. Moreover, increased VEGF‐A expression specifically in the RPE was sufficient to cause choroidal neovascularization (CNV) as in neovascular AMD, which could be inhibited by RPE‐specific inactivation of Flk1, while Tlr2 inactivation strongly reduced CNV. These findings suggest a shared pathogenic role of VEGF‐A‐induced and NLRP3 inflammasome‐mediated IL‐1β activation for multiple distinct ocular aging diseases.
Collapse
Affiliation(s)
- Alexander G Marneros
- Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, MA, USA Department of Dermatology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
174
|
Semeniuk-Wojtaś A, Lubas A, Stec R, Szczylik C, Niemczyk S. Influence of Tyrosine Kinase Inhibitors on Hypertension and Nephrotoxicity in Metastatic Renal Cell Cancer Patients. Int J Mol Sci 2016; 17:ijms17122073. [PMID: 27941701 PMCID: PMC5187873 DOI: 10.3390/ijms17122073] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Revised: 11/06/2016] [Accepted: 12/02/2016] [Indexed: 01/09/2023] Open
Abstract
Renal cell carcinoma (RCC) is one of the most common kidney malignancies. An upgraded comprehension of the molecular biology implicated in the development of cancer has stimulated an increase in research and development of innovative antitumor therapies. The aim of the study was to analyze the medical literature for hypertension and renal toxicities as the adverse events of the vascular endothelial growth factor (VEGF) signaling pathway inhibitor (anti-VEGF) therapy. Relevant studies were identified in PubMed and ClinicalTrials.gov databases. Eligible studies were phase III and IV prospective clinical trials, meta-analyses and retrospective studies that had described events of hypertension or nephrotoxicity for patients who received anti-VEGF therapy. A total of 48 studies were included in the systematic review. The incidence of any grade hypertension ranged from 17% to 49.6%. Proteinuria and increased creatinine levels were ascertained in 8% to 73% and 5% to 65.6% of patients, respectively. These adverse events are most often mild in severity but may sometimes lead to treatment discontinuation. Nephrotoxicity and hypertension are related to multiple mechanisms; however, one of the main disturbances in those patients is VEGF inhibition. There is a significant risk of developing hypertension and renal dysfunction among patients receiving anti-VEGF treatment; however, there is also some evidence that these side effects may be used as biomarkers of response to antiangiogenic agents.
Collapse
Affiliation(s)
| | - Arkadiusz Lubas
- Military Institute of Medicine Szaserów, 128 Street, 04-141 Warsaw, Poland.
| | - Rafał Stec
- Military Institute of Medicine Szaserów, 128 Street, 04-141 Warsaw, Poland.
| | - Cezary Szczylik
- Military Institute of Medicine Szaserów, 128 Street, 04-141 Warsaw, Poland.
| | - Stanisław Niemczyk
- Military Institute of Medicine Szaserów, 128 Street, 04-141 Warsaw, Poland.
| |
Collapse
|
175
|
Rozance PJ, Hay WW. Pancreatic islet hepatocyte growth factor and vascular endothelial growth factor A signaling in growth restricted fetuses. Mol Cell Endocrinol 2016; 435:78-84. [PMID: 26820125 PMCID: PMC4959995 DOI: 10.1016/j.mce.2016.01.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 01/16/2016] [Accepted: 01/22/2016] [Indexed: 12/31/2022]
Abstract
Placental insufficiency leads to intrauterine growth restriction (IUGR) and a lifelong risk of developing type 2 diabetes. Impaired islet development in the growth restricted fetus, including decreased β-cell replication, mass, and insulin secretion, is strongly implicated in the pathogenesis of later life type 2 diabetes. Currently, standard medical management of a woman with a pregnancy complicated by placental insufficiency and fetal IUGR is increased fetal surveillance and indicated preterm delivery. This leads to the dual complications of IUGR and preterm birth - both of which may increase the lifelong risk for type 2 diabetes. In order to develop therapeutic interventions in IUGR pregnancies complicated by placental insufficiency and decrease the risk of later development of type 2 diabetes in the offspring, the mechanisms responsible for impaired islet development in these cases must be determined. This review focuses on current investigations testing the hypothesis that decreased nutrient supply to the IUGR fetus inhibits an intra-islet hepatocyte growth factor - vascular endothelial growth factor A (HGF - VEGFA) feed forward signaling pathway and that this is responsible for developmental islet defects.
Collapse
Affiliation(s)
- Paul J Rozance
- Perinatal Research Center, University of Colorado Denver School of Medicine, Department of Pediatrics, USA.
| | - William W Hay
- Perinatal Research Center, University of Colorado Denver School of Medicine, Department of Pediatrics, USA
| |
Collapse
|
176
|
Baghi A, Jabbarpoor Bonyadi MH, Ramezani A, Azarmina M, Moradian S, Dehghan MH, Nourinia R, Peyman GA, Yaseri M, Soheilian M. Two Doses of Intravitreal Ziv-Aflibercept versus Bevacizumab in Treatment of Diabetic Macular Edema: A Three-Armed, Double-Blind Randomized Trial. Ophthalmol Retina 2016; 1:103-110. [PMID: 31047266 DOI: 10.1016/j.oret.2016.08.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 08/01/2016] [Accepted: 08/01/2016] [Indexed: 01/29/2023]
Abstract
PURPOSE To compare the efficacy of 2 doses of intravitreal ziv-aflibercept (IVZ) with intravitreal bevacizumab (IVB) in the treatment of center-involved diabetic macular edema (DME) at 12 weeks. DESIGN Three-armed, double-blind, randomized clinical trial. PARTICIPANTS Eyes with center-involved DME. METHODS In this trial, 123 eyes with DME were randomly assigned to 3 injections of 1.25 mg IVZ, 2.5 mg IVZ, and 1.25 mg IVB every 4 weeks. Complete ophthalmologic examination and central macular thickness (CMT) measurement by optical coherence tomography were performed every 4 weeks up to 12 weeks. MAIN OUTCOME MEASURES Change in best-corrected visual acuity (BCVA) at 12 weeks. RESULTS Although no significant difference was evident between the 2 ziv-aflibercept groups at 12 weeks, the BCVA change was significantly better in the ziv-aflibercept 1.25 mg group than in the IVB group at the 12-week visit (P = 0.021). In regard to CMT changes, there was no significant difference between the 2 ziv-aflibercept groups; however, a significantly greater reduction in CMT was observed in the ziv-aflibercept 2.5 mg group compared with the IVB group at 12 weeks (P = 0.037). Subgroup analysis disclosed no difference in BCVA outcomes at 12 weeks among the groups in the eyes with baseline BCVA ≥20/50. In the eyes with baseline BCVA <20/50, the improvement was significantly better at 12 weeks in the ziv-aflibercept 1.25 mg group compared with the IVB group (P = 0.011). CONCLUSIONS The 12-week results of this trial disclosed that both 1.25 mg and 2.5 mg doses of IVZ and IVB demonstrated BCVA improvement over baseline in the treatment of center-involved DME. However, a stronger effect of IVZ compared with IVB in terms of both visual acuity improvement and macular thickness reduction was detected in the eyes with initial BCVA <20/50. Longer-term efficacy and safety data will be needed to understand the role for this drug in practice.
Collapse
Affiliation(s)
- Ahmadreza Baghi
- Ophthalmology Department and Ophthalmic Research Center, Labbafinejad Medical Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Jabbarpoor Bonyadi
- Ophthalmology Department and Ophthalmic Research Center, Labbafinejad Medical Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Ramezani
- Ophthalmology Department and Ophthalmic Research Center, Labbafinejad Medical Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Negah Eye Hospital, Tehran, Iran; Torfe Medical Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Azarmina
- Ophthalmology Department and Ophthalmic Research Center, Labbafinejad Medical Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Negah Eye Hospital, Tehran, Iran
| | - Siamak Moradian
- Ophthalmology Department and Ophthalmic Research Center, Labbafinejad Medical Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Negah Eye Hospital, Tehran, Iran
| | - Mohammad Hossein Dehghan
- Ophthalmology Department and Ophthalmic Research Center, Labbafinejad Medical Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Negah Eye Hospital, Tehran, Iran
| | - Ramin Nourinia
- Ophthalmology Department and Ophthalmic Research Center, Labbafinejad Medical Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Gholam A Peyman
- Department of Ophthalmology and Basic Medical Sciences, University of Arizona, Phoenix, Arizona
| | - Mehdi Yaseri
- Ophthalmology Department and Ophthalmic Research Center, Labbafinejad Medical Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoud Soheilian
- Ophthalmology Department and Ophthalmic Research Center, Labbafinejad Medical Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Negah Eye Hospital, Tehran, Iran.
| |
Collapse
|
177
|
Abstract
With the incorporation of targeted therapies in routine cancer therapy, it is imperative that the array of toxicities associated with these agents be well-recognized and managed, especially since these toxicities are distinct from those seen with conventional cytotoxic agents. This review will focus on these renal toxicities from commonly used targeted agents. This review discusses the mechanisms of these side effects and management strategies. Anti-vascular endothelial growth factor (VEGF) agents including the monoclonal antibody bevacizumab, aflibercept (VEGF trap), and anti-VEGF receptor (VEGFR) tyrosine kinase inhibitors (TKIs) all cause hypertension, whereas some of them result in proteinuria. Monoclonal antibodies against the human epidermal growth factor receptor (HER) family of receptors, such as cetuximab and panitumumab, cause electrolyte imbalances including hypomagnesemia and hypokalemia due to the direct nephrotoxic effect of the drug on renal tubules. Cetuximab may also result in renal tubular acidosis. The TKIs, imatinib and dasatinib, can result in acute or chronic renal failure. Rituximab, an anti-CD20 monoclonal antibody, can cause acute renal failure following initiation of therapy because of the onset of acute tumor lysis syndrome. Everolimus, a mammalian target of rapamycin (mTOR) inhibitor, can result in proteinuria. Discerning the renal adverse effects resulting from these agents is essential for safe treatment strategies, particularly in those with pre-existing renal disease.
Collapse
Affiliation(s)
- Anum Abbas
- Department of Internal Medicine, School of Medicine, Creighton University, Omaha, NE, USA
| | - Mohsin M Mirza
- Department of Internal Medicine, School of Medicine, Creighton University, Omaha, NE, USA
| | - Apar Kishor Ganti
- Division of Oncology and Hematology, Department of Internal Medicine, VA-Nebraska Western Iowa Health Care System and University of Nebraska Medical Center, Omaha, NE, USA
| | - Ketki Tendulkar
- Division of Nephrology, Department of Internal Medicine, University of Nebraska Medical Center, 983040 Nebraska Medical Center, Omaha, NE, 68198-3040, USA.
| |
Collapse
|
178
|
Systemic safety of intravitreal anti-vascular endothelial growth factor agents in age-related macular degeneration. Curr Opin Ophthalmol 2016; 27:224-43. [PMID: 26871657 DOI: 10.1097/icu.0000000000000257] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
PURPOSE OF REVIEW The purpose of review is to summarize the literature addressing nonocular adverse events in patients with neovascular age-related macular degeneration treated with intravitreal vascular endothelial growth factor (VEGF) inhibitors and to present possible mechanisms of effect. RECENT FINDINGS The incidence of overall nonocular serious adverse events varied from 0 to 39.3% and nonocular adverse events ranged from 0 to 86.9%. Few studies have reported a significant association between use of intravitreal anti-VEGF agents and overall incidence of adverse events, stroke, myocardial infarction, nonocular hemorrhage and death, with overall greater concern in patients treated with bevacizumab. Additionally, history of stroke or other arterial thromboembolic event may be a risk factor for future stroke in patients treated with intravitreal anti-VEGF agents. Theories explaining the mechanisms of increased risk of nonocular adverse events secondary to anti-VEGF agent use surround the necessity of VEGF for the normal functioning of the endothelium and the damage incurred with use of anti-VEGF agents. SUMMARY Current data are insufficient to definitively conclude that intravitreal anti-VEGF agents are safe, although there is a trend toward an overall favorable systemic safety profile. Caution should be exerted in patients with a history of cardiovascular disease, as these patients may be at greater risk for nonocular serious adverse events.
Collapse
|
179
|
Bábíčková J, Klinkhammer BM, Buhl EM, Djudjaj S, Hoss M, Heymann F, Tacke F, Floege J, Becker JU, Boor P. Regardless of etiology, progressive renal disease causes ultrastructural and functional alterations of peritubular capillaries. Kidney Int 2016; 91:70-85. [PMID: 27678159 DOI: 10.1016/j.kint.2016.07.038] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 07/15/2016] [Accepted: 07/28/2016] [Indexed: 12/28/2022]
Abstract
Progressive renal diseases are associated with rarefaction of peritubular capillaries, but the ultrastructural and functional alterations of the microvasculature are not well described. To study this, we analyzed different time points during progressive kidney damage and fibrosis in 3 murine models of different disease etiologies. These models were unilateral ureteral obstruction, unilateral ischemia-reperfusion injury, and Col4a3-deficient mice, we analyzed ultrastructural alterations in patient biopsy specimens. Compared with kidneys of healthy mice, we found a significant and progressive reduction of peritubular capillaries in all models analyzed. Ultrastructurally, compared with the kidneys of control mice, focal widening of the subendothelial space and higher numbers of endothelial vacuoles and caveolae were found in fibrotic kidneys. Quantitative analysis showed that peritubular capillary endothelial cells in fibrotic kidneys had significantly and progressively reduced numbers of fenestrations and increased thickness of the cell soma and lamina densa of the capillary basement membrane. Similar ultrastructural changes were also observed in patient's kidney biopsy specimens. Compared with healthy murine kidneys, fibrotic kidneys had significantly increased extravasation of Evans blue dye in all 3 models. The extravasation could be visualized using 2-photon microscopy in real time in living animals and was mainly localized to capillary branching points. Finally, fibrotic kidneys in all models exhibited a significantly greater degree of interstitial deposition of fibrinogen. Thus, peritubular capillaries undergo significant ultrastructural and functional alterations during experimental progressive renal diseases, independent of the underlying injury. Analyses of these alterations could provide read-outs for the evaluation of therapeutic approaches targeting the renal microvasculature.
Collapse
Affiliation(s)
- Janka Bábíčková
- Institute of Pathology, RWTH University of Aachen, Aachen, Germany; Division of Nephrology, RWTH University of Aachen, Aachen, Germany; Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia; Institute for Clinical and Translational Research, Biomedical Research Center SAS, Bratislava, Slovakia
| | | | - Eva M Buhl
- Institute of Pathology, RWTH University of Aachen, Aachen, Germany; Division of Nephrology, RWTH University of Aachen, Aachen, Germany
| | - Sonja Djudjaj
- Institute of Pathology, RWTH University of Aachen, Aachen, Germany
| | - Mareike Hoss
- Institute of Pathology, RWTH University of Aachen, Aachen, Germany; Electron Microscopy Facility, RWTH University of Aachen, Aachen, Germany
| | - Felix Heymann
- Division of Gastroenterology, RWTH University of Aachen, Aachen, Germany
| | - Frank Tacke
- Division of Gastroenterology, RWTH University of Aachen, Aachen, Germany
| | - Jürgen Floege
- Division of Nephrology, RWTH University of Aachen, Aachen, Germany
| | - Jan U Becker
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Peter Boor
- Institute of Pathology, RWTH University of Aachen, Aachen, Germany; Division of Nephrology, RWTH University of Aachen, Aachen, Germany.
| |
Collapse
|
180
|
Tanaka M, Iwakiri Y. The Hepatic Lymphatic Vascular System: Structure, Function, Markers, and Lymphangiogenesis. Cell Mol Gastroenterol Hepatol 2016; 2:733-749. [PMID: 28105461 PMCID: PMC5240041 DOI: 10.1016/j.jcmgh.2016.09.002] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 09/02/2016] [Indexed: 02/06/2023]
Abstract
The lymphatic vascular system has been minimally explored in the liver despite its essential functions including maintenance of tissue fluid homeostasis. The discovery of specific markers for lymphatic endothelial cells has advanced the study of lymphatics by methods including imaging, cell isolation, and transgenic animal models and has resulted in rapid progress in lymphatic vascular research during the last decade. These studies have yielded concrete evidence that lymphatic vessel dysfunction plays an important role in the pathogenesis of many diseases. This article reviews the current knowledge of the structure, function, and markers of the hepatic lymphatic vascular system as well as factors associated with hepatic lymphangiogenesis and compares liver lymphatics with those in other tissues.
Collapse
Key Words
- CCl4, carbon tetrachloride
- Cirrhosis
- EHE, epithelioid hemangioendothelioma
- HA, hyaluronan
- HBx Ag, hepatitis B x antigen
- HCC, hepatocellular carcinoma
- IFN, interferon
- IL, interleukin
- Inflammation
- LSEC, liver sinusoidal endothelial cell
- LYVE-1, lymphatic vessel endothelial hyaluronan receptor 1
- LyEC, lymphatic endothelial cell
- NO, nitric oxide
- Portal Hypertension
- Prox1, prospero homeobox protein 1
- VEGF
- VEGF, vascular endothelial growth factor
- VEGFR, vascular endothelial growth factor receptor
- mTOR, mammalian target of rapamycin
Collapse
Affiliation(s)
| | - Yasuko Iwakiri
- Reprint requests Address requests for reprints to: Yasuko Iwakiri, PhD, Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, TAC S223B, 333 Cedar Street, New Haven, Connecticut 06520. fax: (203) 785-7273.Section of Digestive DiseasesDepartment of Internal MedicineYale University School of MedicineTAC S223B, 333 Cedar StreetNew HavenConnecticut 06520
| |
Collapse
|
181
|
Block KI, Gyllenhaal C, Lowe L, Amedei A, Amin ARMR, Amin A, Aquilano K, Arbiser J, Arreola A, Arzumanyan A, Ashraf SS, Azmi AS, Benencia F, Bhakta D, Bilsland A, Bishayee A, Blain SW, Block PB, Boosani CS, Carey TE, Carnero A, Carotenuto M, Casey SC, Chakrabarti M, Chaturvedi R, Chen GZ, Chen H, Chen S, Chen YC, Choi BK, Ciriolo MR, Coley HM, Collins AR, Connell M, Crawford S, Curran CS, Dabrosin C, Damia G, Dasgupta S, DeBerardinis RJ, Decker WK, Dhawan P, Diehl AME, Dong JT, Dou QP, Drew JE, Elkord E, El-Rayes B, Feitelson MA, Felsher DW, Ferguson LR, Fimognari C, Firestone GL, Frezza C, Fujii H, Fuster MM, Generali D, Georgakilas AG, Gieseler F, Gilbertson M, Green MF, Grue B, Guha G, Halicka D, Helferich WG, Heneberg P, Hentosh P, Hirschey MD, Hofseth LJ, Holcombe RF, Honoki K, Hsu HY, Huang GS, Jensen LD, Jiang WG, Jones LW, Karpowicz PA, Keith WN, Kerkar SP, Khan GN, Khatami M, Ko YH, Kucuk O, Kulathinal RJ, Kumar NB, Kwon BS, Le A, Lea MA, Lee HY, Lichtor T, Lin LT, Locasale JW, Lokeshwar BL, Longo VD, Lyssiotis CA, MacKenzie KL, Malhotra M, Marino M, Martinez-Chantar ML, Matheu A, Maxwell C, McDonnell E, Meeker AK, Mehrmohamadi M, Mehta K, Michelotti GA, Mohammad RM, Mohammed SI, Morre DJ, Muralidhar V, Muqbil I, Murphy MP, Nagaraju GP, Nahta R, Niccolai E, Nowsheen S, Panis C, Pantano F, Parslow VR, Pawelec G, Pedersen PL, Poore B, Poudyal D, Prakash S, Prince M, Raffaghello L, Rathmell JC, Rathmell WK, Ray SK, Reichrath J, Rezazadeh S, Ribatti D, Ricciardiello L, Robey RB, Rodier F, Rupasinghe HPV, Russo GL, Ryan EP, Samadi AK, Sanchez-Garcia I, Sanders AJ, Santini D, Sarkar M, Sasada T, Saxena NK, Shackelford RE, Shantha Kumara HMC, Sharma D, Shin DM, Sidransky D, Siegelin MD, Signori E, Singh N, Sivanand S, Sliva D, Smythe C, Spagnuolo C, Stafforini DM, Stagg J, Subbarayan PR, Sundin T, Talib WH, Thompson SK, Tran PT, Ungefroren H, Vander Heiden MG, Venkateswaran V, Vinay DS, Vlachostergios PJ, Wang Z, Wellen KE, Whelan RL, Yang ES, Yang H, Yang X, Yaswen P, Yedjou C, Yin X, Zhu J, Zollo M. Designing a broad-spectrum integrative approach for cancer prevention and treatment. Semin Cancer Biol 2016; 35 Suppl:S276-S304. [PMID: 26590477 DOI: 10.1016/j.semcancer.2015.09.007] [Citation(s) in RCA: 190] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 08/12/2015] [Accepted: 09/14/2015] [Indexed: 12/14/2022]
Abstract
Targeted therapies and the consequent adoption of "personalized" oncology have achieved notable successes in some cancers; however, significant problems remain with this approach. Many targeted therapies are highly toxic, costs are extremely high, and most patients experience relapse after a few disease-free months. Relapses arise from genetic heterogeneity in tumors, which harbor therapy-resistant immortalized cells that have adopted alternate and compensatory pathways (i.e., pathways that are not reliant upon the same mechanisms as those which have been targeted). To address these limitations, an international task force of 180 scientists was assembled to explore the concept of a low-toxicity "broad-spectrum" therapeutic approach that could simultaneously target many key pathways and mechanisms. Using cancer hallmark phenotypes and the tumor microenvironment to account for the various aspects of relevant cancer biology, interdisciplinary teams reviewed each hallmark area and nominated a wide range of high-priority targets (74 in total) that could be modified to improve patient outcomes. For these targets, corresponding low-toxicity therapeutic approaches were then suggested, many of which were phytochemicals. Proposed actions on each target and all of the approaches were further reviewed for known effects on other hallmark areas and the tumor microenvironment. Potential contrary or procarcinogenic effects were found for 3.9% of the relationships between targets and hallmarks, and mixed evidence of complementary and contrary relationships was found for 7.1%. Approximately 67% of the relationships revealed potentially complementary effects, and the remainder had no known relationship. Among the approaches, 1.1% had contrary, 2.8% had mixed and 62.1% had complementary relationships. These results suggest that a broad-spectrum approach should be feasible from a safety standpoint. This novel approach has potential to be relatively inexpensive, it should help us address stages and types of cancer that lack conventional treatment, and it may reduce relapse risks. A proposed agenda for future research is offered.
Collapse
Affiliation(s)
- Keith I Block
- Block Center for Integrative Cancer Treatment, Skokie, IL, United States.
| | | | - Leroy Lowe
- Getting to Know Cancer, Truro, Nova Scotia, Canada; Lancaster Environment Centre, Lancaster University, Bailrigg, Lancaster, United Kingdom.
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - A R M Ruhul Amin
- Winship Cancer Institute of Emory University, Atlanta, GA, United States
| | - Amr Amin
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Katia Aquilano
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Jack Arbiser
- Winship Cancer Institute of Emory University, Atlanta, GA, United States; Atlanta Veterans Administration Medical Center, Atlanta, GA, United States; Department of Dermatology, Emory University School of Medicine, Emory University, Atlanta, GA, United States
| | - Alexandra Arreola
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, United States
| | - Alla Arzumanyan
- Department of Biology, Temple University, Philadelphia, PA, United States
| | - S Salman Ashraf
- Department of Chemistry, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Asfar S Azmi
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| | - Fabian Benencia
- Department of Biomedical Sciences, Ohio University, Athens, OH, United States
| | - Dipita Bhakta
- School of Chemical and Bio Technology, SASTRA University, Thanjavur, Tamil Nadu, India
| | | | - Anupam Bishayee
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin Health Sciences Institute, Miami, FL, United States
| | - Stacy W Blain
- Department of Pediatrics, State University of New York, Downstate Medical Center, Brooklyn, NY, United States
| | - Penny B Block
- Block Center for Integrative Cancer Treatment, Skokie, IL, United States
| | - Chandra S Boosani
- Department of BioMedical Sciences, School of Medicine, Creighton University, Omaha, NE, United States
| | - Thomas E Carey
- Head and Neck Cancer Biology Laboratory, University of Michigan, Ann Arbor, MI, United States
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla, Consejo Superior de Investigaciones Cientificas, Seville, Spain
| | - Marianeve Carotenuto
- Centro di Ingegneria Genetica e Biotecnologia Avanzate, Naples, Italy; Department of Molecular Medicine and Medical Biotechnology, Federico II, Via Pansini 5, 80131 Naples, Italy
| | - Stephanie C Casey
- Stanford University, Division of Oncology, Department of Medicine and Pathology, Stanford, CA, United States
| | - Mrinmay Chakrabarti
- Department of Pathology, Microbiology, and Immunology, University of South Carolina, School of Medicine, Columbia, SC, United States
| | - Rupesh Chaturvedi
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Georgia Zhuo Chen
- Winship Cancer Institute of Emory University, Atlanta, GA, United States
| | - Helen Chen
- Department of Pediatrics, University of British Columbia, Michael Cuccione Childhood Cancer Research Program, Child and Family Research Institute, Vancouver, British Columbia, Canada
| | - Sophie Chen
- Ovarian and Prostate Cancer Research Laboratory, Guildford, Surrey, United Kingdom
| | - Yi Charlie Chen
- Department of Biology, Alderson Broaddus University, Philippi, WV, United States
| | - Beom K Choi
- Cancer Immunology Branch, Division of Cancer Biology, National Cancer Center, Goyang, Gyeonggi, Republic of Korea
| | | | - Helen M Coley
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, United Kingdom
| | - Andrew R Collins
- Department of Nutrition, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Marisa Connell
- Department of Pediatrics, University of British Columbia, Michael Cuccione Childhood Cancer Research Program, Child and Family Research Institute, Vancouver, British Columbia, Canada
| | - Sarah Crawford
- Cancer Biology Research Laboratory, Southern Connecticut State University, New Haven, CT, United States
| | - Colleen S Curran
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Charlotta Dabrosin
- Department of Oncology and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Giovanna Damia
- Department of Oncology, Istituto Di Ricovero e Cura a Carattere Scientifico - Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Santanu Dasgupta
- Department of Cellular and Molecular Biology, the University of Texas Health Science Center at Tyler, Tyler, TX, United States
| | - Ralph J DeBerardinis
- Children's Medical Center Research Institute, University of Texas - Southwestern Medical Center, Dallas, TX, United States
| | - William K Decker
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, United States
| | - Punita Dhawan
- Department of Surgery and Cancer Biology, Division of Surgical Oncology, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Anna Mae E Diehl
- Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - Jin-Tang Dong
- Winship Cancer Institute of Emory University, Atlanta, GA, United States
| | - Q Ping Dou
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| | - Janice E Drew
- Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen, Scotland, United Kingdom
| | - Eyad Elkord
- College of Medicine & Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Bassel El-Rayes
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, United States
| | - Mark A Feitelson
- Department of Biology, Temple University, Philadelphia, PA, United States
| | - Dean W Felsher
- Stanford University, Division of Oncology, Department of Medicine and Pathology, Stanford, CA, United States
| | - Lynnette R Ferguson
- Discipline of Nutrition and Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Carmela Fimognari
- Dipartimento di Scienze per la Qualità della Vita Alma Mater Studiorum-Università di Bologna, Rimini, Italy
| | - Gary L Firestone
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, CA, United States
| | - Christian Frezza
- Medical Research Council Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge, United Kingdom
| | - Hiromasa Fujii
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Nara, Japan
| | - Mark M Fuster
- Medicine and Research Services, Veterans Affairs San Diego Healthcare System & University of California, San Diego, CA, United States
| | - Daniele Generali
- Department of Medical, Surgery and Health Sciences, University of Trieste, Trieste, Italy; Molecular Therapy and Pharmacogenomics Unit, Azienda Ospedaliera Istituti Ospitalieri di Cremona, Cremona, Italy
| | - Alexandros G Georgakilas
- Physics Department, School of Applied Mathematics and Physical Sciences, National Technical University of Athens, Athens, Greece
| | - Frank Gieseler
- First Department of Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | | | - Michelle F Green
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, United States
| | - Brendan Grue
- Departments of Environmental Science, Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Gunjan Guha
- School of Chemical and Bio Technology, SASTRA University, Thanjavur, Tamil Nadu, India
| | - Dorota Halicka
- Department of Pathology, New York Medical College, Valhalla, NY, United States
| | | | - Petr Heneberg
- Charles University in Prague, Third Faculty of Medicine, Prague, Czech Republic
| | - Patricia Hentosh
- School of Medical Laboratory and Radiation Sciences, Old Dominion University, Norfolk, VA, United States
| | - Matthew D Hirschey
- Department of Medicine, Duke University Medical Center, Durham, NC, United States; Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, United States
| | - Lorne J Hofseth
- College of Pharmacy, University of South Carolina, Columbia, SC, United States
| | - Randall F Holcombe
- Tisch Cancer Institute, Mount Sinai School of Medicine, New York, NY, United States
| | - Kanya Honoki
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Nara, Japan
| | - Hsue-Yin Hsu
- Department of Life Sciences, Tzu-Chi University, Hualien, Taiwan
| | - Gloria S Huang
- Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, United States
| | - Lasse D Jensen
- Department of Medical and Health Sciences, Linköping University, Linköping, Sweden; Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Wen G Jiang
- Cardiff University School of Medicine, Heath Park, Cardiff, United Kingdom
| | - Lee W Jones
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, United States
| | | | | | - Sid P Kerkar
- Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | | | - Mahin Khatami
- Inflammation and Cancer Research, National Cancer Institute (Retired), National Institutes of Health, Bethesda, MD, United States
| | - Young H Ko
- University of Maryland BioPark, Innovation Center, KoDiscovery, Baltimore, MD, United States
| | - Omer Kucuk
- Winship Cancer Institute of Emory University, Atlanta, GA, United States
| | - Rob J Kulathinal
- Department of Biology, Temple University, Philadelphia, PA, United States
| | - Nagi B Kumar
- Moffitt Cancer Center, University of South Florida College of Medicine, Tampa, FL, United States
| | - Byoung S Kwon
- Cancer Immunology Branch, Division of Cancer Biology, National Cancer Center, Goyang, Gyeonggi, Republic of Korea; Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA, United States
| | - Anne Le
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Michael A Lea
- New Jersey Medical School, Rutgers University, Newark, NJ, United States
| | - Ho-Young Lee
- College of Pharmacy, Seoul National University, South Korea
| | - Terry Lichtor
- Department of Neurosurgery, Rush University Medical Center, Chicago, IL, United States
| | - Liang-Tzung Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jason W Locasale
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| | - Bal L Lokeshwar
- Department of Medicine, Georgia Regents University Cancer Center, Augusta, GA, United States
| | - Valter D Longo
- Andrus Gerontology Center, Division of Biogerontology, University of Southern California, Los Angeles, CA, United States
| | - Costas A Lyssiotis
- Department of Molecular and Integrative Physiology and Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI, United States
| | - Karen L MacKenzie
- Children's Cancer Institute Australia, Kensington, New South Wales, Australia
| | - Meenakshi Malhotra
- Department of Biomedical Engineering, McGill University, Montréal, Canada
| | - Maria Marino
- Department of Science, University Roma Tre, Rome, Italy
| | - Maria L Martinez-Chantar
- Metabolomic Unit, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Technology Park of Bizkaia, Bizkaia, Spain
| | | | - Christopher Maxwell
- Department of Pediatrics, University of British Columbia, Michael Cuccione Childhood Cancer Research Program, Child and Family Research Institute, Vancouver, British Columbia, Canada
| | - Eoin McDonnell
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, United States
| | - Alan K Meeker
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mahya Mehrmohamadi
- Field of Genetics, Genomics, and Development, Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, United States
| | - Kapil Mehta
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Gregory A Michelotti
- Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - Ramzi M Mohammad
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| | - Sulma I Mohammed
- Department of Comparative Pathobiology, Purdue University Center for Cancer Research, West Lafayette, IN, United States
| | - D James Morre
- Mor-NuCo, Inc, Purdue Research Park, West Lafayette, IN, United States
| | - Vinayak Muralidhar
- Harvard-MIT Division of Health Sciences and Technology, Harvard Medical School, Boston, MA, United States; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Irfana Muqbil
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, Wellcome Trust-MRC Building, Hills Road, Cambridge, United Kingdom
| | | | - Rita Nahta
- Winship Cancer Institute of Emory University, Atlanta, GA, United States
| | | | - Somaira Nowsheen
- Medical Scientist Training Program, Mayo Graduate School, Mayo Medical School, Mayo Clinic, Rochester, MN, United States
| | - Carolina Panis
- Laboratory of Inflammatory Mediators, State University of West Paraná, UNIOESTE, Paraná, Brazil
| | - Francesco Pantano
- Medical Oncology Department, University Campus Bio-Medico, Rome, Italy
| | - Virginia R Parslow
- Discipline of Nutrition and Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Graham Pawelec
- Center for Medical Research, University of Tübingen, Tübingen, Germany
| | - Peter L Pedersen
- Departments of Biological Chemistry and Oncology, Member at Large, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Brad Poore
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Deepak Poudyal
- College of Pharmacy, University of South Carolina, Columbia, SC, United States
| | - Satya Prakash
- Department of Biomedical Engineering, McGill University, Montréal, Canada
| | - Mark Prince
- Department of Otolaryngology-Head and Neck, Medical School, University of Michigan, Ann Arbor, MI, United States
| | | | - Jeffrey C Rathmell
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, United States
| | - W Kimryn Rathmell
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, United States
| | - Swapan K Ray
- Department of Pathology, Microbiology, and Immunology, University of South Carolina, School of Medicine, Columbia, SC, United States
| | - Jörg Reichrath
- Center for Clinical and Experimental Photodermatology, Clinic for Dermatology, Venerology and Allergology, The Saarland University Hospital, Homburg, Germany
| | - Sarallah Rezazadeh
- Department of Biology, University of Rochester, Rochester, NY, United States
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy & National Cancer Institute Giovanni Paolo II, Bari, Italy
| | - Luigi Ricciardiello
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - R Brooks Robey
- White River Junction Veterans Affairs Medical Center, White River Junction, VT, United States; Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Francis Rodier
- Centre de Rechercher du Centre Hospitalier de l'Université de Montréal and Institut du Cancer de Montréal, Montréal, Quebec, Canada; Université de Montréal, Département de Radiologie, Radio-Oncologie et Médicine Nucléaire, Montréal, Quebec, Canada
| | - H P Vasantha Rupasinghe
- Department of Environmental Sciences, Faculty of Agriculture and Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Gian Luigi Russo
- Institute of Food Sciences National Research Council, Avellino, Italy
| | - Elizabeth P Ryan
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, United States
| | | | - Isidro Sanchez-Garcia
- Experimental Therapeutics and Translational Oncology Program, Instituto de Biología Molecular y Celular del Cáncer, CSIC-Universidad de Salamanca, Salamanca, Spain
| | - Andrew J Sanders
- Cardiff University School of Medicine, Heath Park, Cardiff, United Kingdom
| | - Daniele Santini
- Medical Oncology Department, University Campus Bio-Medico, Rome, Italy
| | - Malancha Sarkar
- Department of Biology, University of Miami, Miami, FL, United States
| | - Tetsuro Sasada
- Department of Immunology, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Neeraj K Saxena
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Rodney E Shackelford
- Department of Pathology, Louisiana State University, Health Shreveport, Shreveport, LA, United States
| | - H M C Shantha Kumara
- Department of Surgery, St. Luke's Roosevelt Hospital, New York, NY, United States
| | - Dipali Sharma
- Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, United States
| | - Dong M Shin
- Winship Cancer Institute of Emory University, Atlanta, GA, United States
| | - David Sidransky
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Markus David Siegelin
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, United States
| | - Emanuela Signori
- National Research Council, Institute of Translational Pharmacology, Rome, Italy
| | - Neetu Singh
- Advanced Molecular Science Research Centre (Centre for Advanced Research), King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Sharanya Sivanand
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Daniel Sliva
- DSTest Laboratories, Purdue Research Park, Indianapolis, IN, United States
| | - Carl Smythe
- Department of Biomedical Science, Sheffield Cancer Research Centre, University of Sheffield, Sheffield, United Kingdom
| | - Carmela Spagnuolo
- Institute of Food Sciences National Research Council, Avellino, Italy
| | - Diana M Stafforini
- Huntsman Cancer Institute and Department of Internal Medicine, University of Utah, Salt Lake City, UT, United States
| | - John Stagg
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Faculté de Pharmacie et Institut du Cancer de Montréal, Montréal, Quebec, Canada
| | - Pochi R Subbarayan
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Tabetha Sundin
- Department of Molecular Diagnostics, Sentara Healthcare, Norfolk, VA, United States
| | - Wamidh H Talib
- Department of Clinical Pharmacy and Therapeutics, Applied Science University, Amman, Jordan
| | - Sarah K Thompson
- Department of Surgery, Royal Adelaide Hospital, Adelaide, Australia
| | - Phuoc T Tran
- Departments of Radiation Oncology & Molecular Radiation Sciences, Oncology and Urology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Hendrik Ungefroren
- First Department of Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Matthew G Vander Heiden
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Vasundara Venkateswaran
- Department of Surgery, University of Toronto, Division of Urology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Dass S Vinay
- Section of Clinical Immunology, Allergy, and Rheumatology, Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA, United States
| | - Panagiotis J Vlachostergios
- Department of Internal Medicine, New York University Lutheran Medical Center, Brooklyn, New York, NY, United States
| | - Zongwei Wang
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Kathryn E Wellen
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Richard L Whelan
- Department of Surgery, St. Luke's Roosevelt Hospital, New York, NY, United States
| | - Eddy S Yang
- Department of Radiation Oncology, University of Alabama at Birmingham School of Medicine, Birmingham, AL, United States
| | - Huanjie Yang
- The School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang, China
| | - Xujuan Yang
- University of Illinois at Urbana Champaign, Champaign, IL, United States
| | - Paul Yaswen
- Life Sciences Division, Lawrence Berkeley National Lab, Berkeley, CA, United States
| | - Clement Yedjou
- Department of Biology, Jackson State University, Jackson, MS, United States
| | - Xin Yin
- Medicine and Research Services, Veterans Affairs San Diego Healthcare System & University of California, San Diego, CA, United States
| | - Jiyue Zhu
- Washington State University College of Pharmacy, Spokane, WA, United States
| | - Massimo Zollo
- Centro di Ingegneria Genetica e Biotecnologia Avanzate, Naples, Italy; Department of Molecular Medicine and Medical Biotechnology, Federico II, Via Pansini 5, 80131 Naples, Italy
| |
Collapse
|
182
|
Turner RJ, Eikmans M, Bajema IM, Bruijn JA, Baelde HJ. Stability and Species Specificity of Renal VEGF-A Splicing Patterns in Kidney Disease. PLoS One 2016; 11:e0162166. [PMID: 27598902 PMCID: PMC5012578 DOI: 10.1371/journal.pone.0162166] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 07/08/2016] [Indexed: 12/29/2022] Open
Abstract
Vascular endothelial growth factor A (VEGF-A) is essential for maintaining the glomerular filtration barrier. Absolute renal levels of VEGF-A change in patients with diabetic nephropathy and inflammatory kidney diseases, but whether changes in the renal splicing patterns of VEGF-A play a role remains unclear. In this study, we investigated mRNA splicing patterns of pro-angiogenic isoforms of VEGF-A in glomeruli and whole kidney samples from human patients with kidney disease and from mouse models of kidney disease. Kidney biopsies were obtained from patients with acute rejection following kidney transplantation, patients with diabetic nephropathy, and control subjects. In addition, kidney samples were obtained from mice with lupus nephritis, mice with diabetes mellitus, and control mice. The relative expression of each VEGF-A splice variant was measured using RT-PCR followed by quantitative fragment analysis. The pattern of renal VEGF-A splice variants was unchanged in diabetic nephropathy and lupus nephritis and was stable throughout disease progression in acute transplant rejection and diabetic nephropathy; these results suggest renal VEGF-A splicing stability during kidney disease. The splicing patterns were species-specific; in the control human kidney samples, VEGF-A 121 was the dominant isoform, whereas VEGF-A 164 was the dominant isoform measured in the mouse kidney samples.
Collapse
Affiliation(s)
- R. J. Turner
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
- * E-mail:
| | - M. Eikmans
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - I. M. Bajema
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - J. A. Bruijn
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - H. J. Baelde
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
183
|
Discontinuation of anti-VEGF cancer therapy promotes metastasis through a liver revascularization mechanism. Nat Commun 2016; 7:12680. [PMID: 27580750 PMCID: PMC5025794 DOI: 10.1038/ncomms12680] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 07/21/2016] [Indexed: 12/21/2022] Open
Abstract
The impact of discontinuation of anti-VEGF cancer therapy in promoting cancer metastasis is unknown. Here we show discontinuation of anti-VEGF treatment creates a time-window of profound structural changes of liver sinusoidal vasculatures, exhibiting hyper-permeability and enlarged open-pore sizes of the fenestrated endothelium and loss of VE-cadherin. The drug cessation caused highly leaky hepatic vasculatures permit tumour cell intravasation and extravasation. Discontinuation of an anti-VEGF antibody-based drug and sunitinib markedly promotes liver metastasis. Mechanistically, host hepatocyte, but not tumour cell-derived vascular endothelial growth factor (VEGF), is responsible for cancer metastasis. Deletion of hepatocyte VEGF markedly ablates the ‘off-drug'-induced metastasis. These findings provide mechanistic insights on anti-VEGF cessation-induced metastasis and raise a new challenge for uninterrupted and sustained antiangiogenic therapy for treatment of human cancers. Anti-VEGF therapy often produces limited beneficial effects in cancer patients. Here, the authors show that discontinuation of anti-VEGF cancer therapy in xenografts-bearing mice increases cancer cells extravasation and intravasation in liver through the host-derived VEGF.
Collapse
|
184
|
Factors regulating capillary remodeling in a reversible model of inflammatory corneal angiogenesis. Sci Rep 2016; 6:32137. [PMID: 27561355 PMCID: PMC4999823 DOI: 10.1038/srep32137] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 08/03/2016] [Indexed: 02/06/2023] Open
Abstract
Newly formed microcapillary networks arising in adult organisms by angiogenic and inflammatory stimuli contribute to pathologies such as corneal and retinal blindness, tumor growth, and metastasis. Therapeutic inhibition of pathologic angiogenesis has focused on targeting the VEGF pathway, while comparatively little attention has been given to remodeling of the new microcapillaries into a stabilized, functional, and persistent vascular network. Here, we used a novel reversible model of inflammatory angiogenesis in the rat cornea to investigate endogenous factors rapidly invoked to remodel, normalize and regress microcapillaries as part of the natural response to regain corneal avascularity. Rapid reversal of an inflammatory angiogenic stimulus suppressed granulocytic activity, enhanced recruitment of remodelling macrophages, induced capillary intussusception, and enriched pathways and processes involving immune cells, chemokines, morphogenesis, axonal guidance, and cell motility, adhesion, and cytoskeletal functions. Whole transcriptome gene expression analysis revealed suppression of numerous inflammatory and angiogenic factors and enhancement of endogenous inhibitors. Many of the identified genes function independently of VEGF and represent potentially new targets for molecular control of the critical process of microvascular remodeling and regression in the cornea.
Collapse
|
185
|
Nurmi H, Saharinen P, Zarkada G, Zheng W, Robciuc MR, Alitalo K. VEGF-C is required for intestinal lymphatic vessel maintenance and lipid absorption. EMBO Mol Med 2016; 7:1418-25. [PMID: 26459520 PMCID: PMC4644375 DOI: 10.15252/emmm.201505731] [Citation(s) in RCA: 169] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Vascular endothelial growth factor C (VEGF-C) binding to its tyrosine kinase receptor VEGFR-3 drives lymphatic vessel growth during development and in pathological processes. Although the VEGF-C/VEGFR-3 pathway provides a target for treatment of cancer and lymphedema, the physiological functions of VEGF-C in adult vasculature are unknown. We show here that VEGF-C is necessary for perinatal lymphangiogenesis, but required for adult lymphatic vessel maintenance only in the intestine. Following Vegfc gene deletion in adult mice, the intestinal lymphatic vessels, including the lacteal vessels, underwent gradual atrophy, which was aggravated when also Vegfd was deleted. VEGF-C was expressed by a subset of smooth muscle cells adjacent to the lacteals in the villus and in the intestinal wall. The Vegfc-deleted mice showed defective lipid absorption and increased fecal excretion of dietary cholesterol and fatty acids. When fed a high-fat diet, the Vegfc-deficient mice were resistant to obesity and had improved glucose metabolism. Our findings indicate that the lymphangiogenic growth factors provide trophic and dynamic regulation of the intestinal lymphatic vasculature, which could be especially important in the dietary regulation of adiposity and cholesterol metabolism.
Collapse
Affiliation(s)
- Harri Nurmi
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki University of Helsinki, Helsinki, Finland
| | - Pipsa Saharinen
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki University of Helsinki, Helsinki, Finland
| | - Georgia Zarkada
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki University of Helsinki, Helsinki, Finland
| | - Wei Zheng
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki University of Helsinki, Helsinki, Finland
| | - Marius R Robciuc
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki University of Helsinki, Helsinki, Finland
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki University of Helsinki, Helsinki, Finland
| |
Collapse
|
186
|
Bernier-Latmani J, Petrova TV. High-resolution 3D analysis of mouse small-intestinal stroma. Nat Protoc 2016; 11:1617-29. [PMID: 27560169 DOI: 10.1038/nprot.2016.092] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Here we detail a protocol for whole-mount immunostaining of mouse small-intestinal villi that can be used to generate high-resolution 3D images of all gut cell types, including blood and lymphatic vessel cells, neurons, smooth muscle cells, fibroblasts and immune cells. The procedure describes perfusion, fixation, dissection, immunostaining, mounting, clearing, confocal imaging and quantification, using intestinal vasculature as an example. As intestinal epithelial cells prevent visualization with some antibodies, we also provide an optional protocol to remove these cells before fixation. In contrast to alternative current techniques, our protocol enables the entire villus to be visualized with increased spatial resolution of cell location, morphology and cell-cell interactions, thus allowing for easy quantification of phenotypes. The technique, which takes 7 d from mouse dissection to microscopic examination, will be useful for researchers who are interested in most aspects of intestinal biology, including mucosal immunology, infection, nutrition, cancer biology and intestinal microbiota.
Collapse
Affiliation(s)
- Jeremiah Bernier-Latmani
- Department of Fundamental Oncology, Ludwig Institute for Cancer Research and Institute of Pathology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Tatiana V Petrova
- Department of Fundamental Oncology, Ludwig Institute for Cancer Research and Institute of Pathology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences. Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
187
|
Wang Z, Zhang J, Zhang L, Liu P, Xie Y, Zhou Q. Risk of gastrointestinal perforation in cancer patients receiving ramucirumab: a meta-analysis of randomized controlled trials. J Chemother 2016; 28:328-34. [PMID: 26099278 DOI: 10.1179/1973947815y.0000000053] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE Although existing evidence from clinical trials has demonstrated manifestation of gastrointestinal perforation with the use of ramucirumab, overall risks have yet to be reported. Therefore, we performed a meta-analysis of published randomized controlled trials (RCTs) to get a better understanding of the overall incidence and risk of gastrointestinal perforation associated with ramucirumab. METHODS The PubMed and Web of Science databases as well as abstracts presented at American Society of Clinical Oncology conferences were searched to identify relevant studies published up to 01 May 2015. Eligible studies included randomized trials of ramucirumab either alone or in combination with another agent compared with the control arm without ramucirumab and that reported gastrointestinal perforation event. Overall incidence, relative risk (RR) and 95% confidence intervals (CI) were computed using fixed- or random-effects models depending on the heterogeneity of the included studies. RESULTS A total of 4579 patients with a variety of solid malignancies from six RCTs were included in our meta-analysis. The incidence of gastrointestinal perforation related to ramucirumab was 1.5% (95% CI 1.1-2.1%) with a mortality of 29.8% (95% CI 14.9-50.7%). The RR of gastrointestinal perforation associated with ramucirumab was 2.56 (95% CI 1.29-5.09; P = 0.007). CONCLUSIONS Treatment with the ramucirumab is associated with a significant increase in risk of gastrointestinal perforation in cancer patients.
Collapse
Affiliation(s)
- Zexing Wang
- a Department of Oncology , Wuhu No. 2 People's Hospital Affiliated to Wannan Medical College , Anhui Province , China
| | - Jun Zhang
- a Department of Oncology , Wuhu No. 2 People's Hospital Affiliated to Wannan Medical College , Anhui Province , China
| | - Liang Zhang
- b Department of Urology and Institute of Prostatic Diseases , Wuhu No. 2 People's Hospital Affiliated to Wannan Medical College , Anhui Province , China
| | - Pengying Liu
- a Department of Oncology , Wuhu No. 2 People's Hospital Affiliated to Wannan Medical College , Anhui Province , China
| | - Yamin Xie
- a Department of Oncology , Wuhu No. 2 People's Hospital Affiliated to Wannan Medical College , Anhui Province , China
| | - Qin Zhou
- a Department of Oncology , Wuhu No. 2 People's Hospital Affiliated to Wannan Medical College , Anhui Province , China
| |
Collapse
|
188
|
Falcon BL, Chintharlapalli S, Uhlik MT, Pytowski B. Antagonist antibodies to vascular endothelial growth factor receptor 2 (VEGFR-2) as anti-angiogenic agents. Pharmacol Ther 2016; 164:204-25. [PMID: 27288725 DOI: 10.1016/j.pharmthera.2016.06.001] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Interaction of numerous signaling pathways in endothelial and mesangial cells results in exquisite control of the process of physiological angiogenesis, with a central role played by vascular endothelial growth factor receptor 2 (VEGFR-2) and its cognate ligands. However, deregulated angiogenesis participates in numerous pathological processes. Excessive activation of VEGFR-2 has been found to mediate tissue-damaging vascular changes as well as the induction of blood vessel expansion to support the growth of solid tumors. Consequently, therapeutic intervention aimed at inhibiting the VEGFR-2 pathway has become a mainstay of treatment in cancer and retinal diseases. In this review, we introduce the concepts of physiological and pathological angiogenesis, the crucial role played by the VEGFR-2 pathway in these processes, and the various inhibitors of its activity that have entered the clinical practice. We primarily focus on the development of ramucirumab, the antagonist monoclonal antibody (mAb) that inhibits VEGFR-2 and has recently been approved for use in patients with gastric, colorectal, and lung cancers. We examine in-depth the pre-clinical studies using DC101, the mAb to mouse VEGFR-2, which provided a conceptual foundation for the role of VEGFR-2 in physiological and pathological angiogenesis. Finally, we discuss further clinical development of ramucirumab and the future of targeting the VEGF pathway for the treatment of cancer.
Collapse
|
189
|
Karovic S, Shiuan EF, Zhang SQ, Cao H, Maitland ML. Patient-Level Adverse Event Patterns in a Single-Institution Study of the Multi-Kinase Inhibitor Sorafenib. Clin Transl Sci 2016; 9:260-266. [PMID: 27443985 PMCID: PMC5350995 DOI: 10.1111/cts.12408] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 05/27/2016] [Indexed: 01/05/2023] Open
Abstract
Novel characterization of patterns of adverse events (AEs) of kinase inhibitors (KIs) could reveal new insights on human molecular physiology and methods to improve the therapeutic index of KIs. Incidence and severity of AEs for each of 157 patients enrolled in sorafenib clinical trials were determined for three clinically relevant treatment intervals: weeks 0–3, weeks 3–7, and after 7 weeks. The most common within patient co‐occurrences were mucositis with dermatologic events: hand‐foot syndrome (HFS; odds ratio [OR] = 4.36; p = 0.0017) and rash (OR = 5.32; p < 0.001). Prevalence of severe: alopecia (p = 0.02), diarrhea (p < 0.001), and fatigue (p = 0.005) increased over the course of therapy. Incidence of HFS (60%) and diarrhea (25%) increased up to a minimum steady‐state concentration (approximately 5 mcg mL‐1) and plateaued thereafter. Common AEs of sorafenib occur in distinct temporal and tissue distribution patterns and this analysis identified unrecognized relationships among mechanism‐dependent and independent effects of a KI.
Collapse
Affiliation(s)
- S Karovic
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - E F Shiuan
- Department of Biochemistry & Molecular Biology, University of Chicago, Chicago, Illinois, USA
| | - S Q Zhang
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - H Cao
- Department of Health Studies, University of Chicago, Chicago, Illinois, USA
| | - M L Maitland
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois, USA.,Committee on Clinical Pharmacology and Pharmacogenomics, University of Chicago, Chicago, Illinois, USA.,Comprehensive Cancer Center, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
190
|
Gass-Jégu F, Gschwend A, Gairard-Dory AC, Mennecier B, Tebacher-Alt M, Gourieux B, Quoix É. Gastrointestinal perforations in patients treated with erlotinib: A report of two cases with fatal outcome and literature review. Lung Cancer 2016; 99:76-8. [PMID: 27565918 DOI: 10.1016/j.lungcan.2016.06.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 06/11/2016] [Indexed: 11/17/2022]
Abstract
Erlotinib has been approved as second-line treatment in patients with non-small cell lung cancer (NSCLC) experiencing relapse after first-line platinum-based chemotherapy. Herein, we report two occurrences of erlotinib-associated gastrointestinal perforation (GIP) in NSCLC patients. Two patients aged 60 and 79 years received erlotinib as third- and second-line NSCLC treatment, respectively. GIP occurred following 3 weeks and 6 months of erlotinib treatment, leading to death a few days later in both patients, neither of whom had any intestinal metastasis. Risk factors related to erlotinib-induced GIP were concomitant oral corticosteroid therapy and ciprofloxacin administration, which may result in erlotinib overexposure. GIP is a severe adverse drug reaction of erlotinib, infrequently described in the literature, compared to other targeted therapies. The lethal risk of erlotinib-associated GIP should be taken into account when evaluating the benefit-risk balance of erlotinib in patients without epidermal growth factor receptor activating mutations.
Collapse
Affiliation(s)
- Florence Gass-Jégu
- Pharmacy-Sterilisation Department, Strasbourg University Hospital, 1 place de l'hôpital, 67091 Strasbourg cedex, France
| | - Anthony Gschwend
- Chest Disease Department, Strasbourg University Hospital, 1 place de l'hôpital, 67091 Strasbourg cedex, France
| | - Anne-Cécile Gairard-Dory
- Pharmacy-Sterilisation Department, Strasbourg University Hospital, 1 place de l'hôpital, 67091 Strasbourg cedex, France
| | - Bertrand Mennecier
- Chest Disease Department, Strasbourg University Hospital, 1 place de l'hôpital, 67091 Strasbourg cedex, France
| | - Martine Tebacher-Alt
- Pharmacovigilance Department, Strasbourg University Hospital, 1 place de l'hôpital, 67091 Strasbourg cedex, France
| | - Bénédicte Gourieux
- Pharmacy-Sterilisation Department, Strasbourg University Hospital, 1 place de l'hôpital, 67091 Strasbourg cedex, France
| | - Élisabeth Quoix
- Chest Disease Department, Strasbourg University Hospital, 1 place de l'hôpital, 67091 Strasbourg cedex, France.
| |
Collapse
|
191
|
Bozkurt O, Karaca H, Hacıbekiroglu I, Kaplan MA, Duzkopru Y, Uysal M, Berk V, Inanc M, Duran AO, Ozaslan E, Ucar M, Ozkan M. Is sunitinib-induced hypothyroidism a predictive clinical marker for better response in metastatic renal cell carcinoma patients? J Chemother 2016; 28:230-4. [DOI: 10.1179/1973947815y.0000000039] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
192
|
Knapp AE, Goldberg D, Delavar H, Trisko BM, Tang K, Hogan MC, Wagner PD, Breen EC. Skeletal myofiber VEGF regulates contraction-induced perfusion and exercise capacity but not muscle capillarity in adult mice. Am J Physiol Regul Integr Comp Physiol 2016; 311:R192-9. [PMID: 27225953 DOI: 10.1152/ajpregu.00533.2015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 05/17/2016] [Indexed: 11/22/2022]
Abstract
A single bout of exhaustive exercise signals expression of vascular endothelial growth factor (VEGF) in the exercising muscle. Previous studies have reported that mice with life-long deletion of skeletal myofiber VEGF have fewer capillaries and a severe reduction in endurance exercise. However, in adult mice, VEGF gene deletion conditionally targeted to skeletal myofibers limits exercise capacity without evidence of capillary regression. To explain this, we hypothesized that adult skeletal myofiber VEGF acutely regulates skeletal muscle perfusion during muscle contraction. A tamoxifen-inducible skeletal myofiber-specific VEGF gene deletion mouse (skmVEGF-/-) was used to reduce skeletal muscle VEGF protein by 90% in adult mice. Three weeks after inducing deletion of the skeletal myofiber VEGF gene, skmVEGF-/- mice exhibited diminished maximum running speed (-10%, P < 0.05) and endurance capacity (-47%; P < 0.05), which did not persist after 8 wk. In skmVEGF-/- mice, gastrocnemius complex time to fatigue measured in situ was 71% lower than control mice. Contraction-induced perfusion measured by optical imaging during a period of electrically stimulated muscle contraction was 85% lower in skmVEGF-/- than control mice. No evidence of capillary rarefication was detected in the soleus, gastrocnemius, and extensor digitorum longus (EDL) up to 8 wk after tamoxifen-induced VEGF ablation, and contractility and fatigue resistance of the soleus measured ex vivo were also unchanged. The force-frequency of the EDL showed a small right shift, but fatigue resistance did not differ between EDL from control and skmVEGF-/- mice. These data suggest myofiber VEGF is required for regulating perfusion during periods of contraction and may in this manner affect endurance capacity.
Collapse
Affiliation(s)
- Amy E Knapp
- Department of Medicine, University of California, San Diego, La Jolla, California
| | - Daniel Goldberg
- Department of Medicine, University of California, San Diego, La Jolla, California
| | - Hamid Delavar
- Department of Medicine, University of California, San Diego, La Jolla, California
| | - Breanna M Trisko
- Department of Medicine, University of California, San Diego, La Jolla, California
| | - Kechun Tang
- Department of Medicine, University of California, San Diego, La Jolla, California
| | - Michael C Hogan
- Department of Medicine, University of California, San Diego, La Jolla, California
| | - Peter D Wagner
- Department of Medicine, University of California, San Diego, La Jolla, California
| | - Ellen C Breen
- Department of Medicine, University of California, San Diego, La Jolla, California
| |
Collapse
|
193
|
van den Meiracker AH, Danser AHJ. Mechanisms of Hypertension and Renal Injury During Vascular Endothelial Growth Factor Signaling Inhibition. Hypertension 2016; 68:17-23. [PMID: 27185750 DOI: 10.1161/hypertensionaha.116.07618] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Anton H van den Meiracker
- From the Division of Pharmacology and Cardiovascular Medicine, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands.
| | - A H Jan Danser
- From the Division of Pharmacology and Cardiovascular Medicine, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
194
|
Hall AP, Mitchard T, Rolf MG, Stewart J, Duffy P. Femoral Head Growth Plate Dysplasia and Fracture in Juvenile Rabbits Induced by Off-target Antiangiogenic Treatment. Toxicol Pathol 2016; 44:866-73. [PMID: 27162053 DOI: 10.1177/0192623316646483] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Epiphyseal growth plate dysplasia (chondrodysplasia) might be considered as the pathognomonic feature of antiangiogenic treatment in preclinical species as it is reliably and dose-responsively induced in rodents and monkeys with vascular endothelial growth factor receptor (VEGFR) inhibitors, fibroblast growth factor (FGF) receptor inhibitors, matrix metalloproteinase inhibitors, and vascular targeting agents. Here we report epiphyseal growth plate dysplasia in juvenile rabbits treated with an oral spleen tyrosine kinase inhibitor induced by off-target antiangiogenic inhibition of VEGF and FGF family kinase receptors. Epiphyseal growth plate dysplasia resulted in weakening and fracturing of the femoral head physis in 6 of 10 male and 1 of 10 female animals as well as microfracturing and dysplasia of the distal femoral articular cartilage in 1 male animal. Fracture lines ran through the zone of hypertrophic cartilage (as well as adjacent zones), were orientated parallel to the physeal plane, and often involved displacement of the femoral head. We would suggest that the high prevalence of growth plate fracture in the rabbit may represent a potential additional adverse risk to those already established for children treated with antiangiogenic therapy.
Collapse
Affiliation(s)
- A Peter Hall
- AstraZeneca, Drug Safety and Metabolism, Macclesfield, Cheshire, UK
| | - T Mitchard
- AstraZeneca, Drug Safety and Metabolism, Macclesfield, Cheshire, UK
| | - M G Rolf
- AstraZeneca, Drug Safety and Metabolism, Pepparedsleden, Gothenburg, Sweden
| | - J Stewart
- AstraZeneca, Drug Safety and Metabolism, Macclesfield, Cheshire, UK
| | - P Duffy
- AstraZeneca, Drug Safety and Metabolism, Macclesfield, Cheshire, UK
| |
Collapse
|
195
|
Örnek N, Örnek K, Aydin S, Yilmaz M, Ölmez Y. Serum vascular endothelial growth factor receptor-2 and adropin levels in age-related macular degeneration. Int J Ophthalmol 2016; 9:556-60. [PMID: 27162728 DOI: 10.18240/ijo.2016.04.13] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 07/20/2015] [Indexed: 12/16/2022] Open
Abstract
AIM To investigate the serum levels of vascular endothelial growth factor receptor-2 (VEGFR-2) and adropin in age-related macular degeneration (AMD) patients. METHODS Ninety-eight AMD patients were included in the study. Seventy-eight age- and sex-matched healthy volunteers were recruited as the control group. Fundus florescein angiography and optical coherence tomography were performed to assess the posterior segment details. Serum VEGFR-2 and adropin levels were measured using enzyme-linked immunosorbent assays and compared between the study groups. RESULTS AMD group had significantly increased foveal retinal thickness, serum LDL and HDL levels and significantly decreased subfoveal choroidal thickness (P =0.01, 0.047, 0.025 and <0.001, respectively). Serum VEGFR-2 level revealed a significant decrease in AMD patients compared to controls (26.48±6.44 vs 30.42±7.92 ng/mL, P<0.001). There was an insignificant increase in serum adropin level in AMD patients (6.17±3.19 vs 5.79±2.71 ng/mL, P=0.4). Serum level of VEGFR-2 in AMD patients had a significant negative correlation with foveal retinal thickness (r=-0.226, P=0.025) and a significant positive correlation with subfoveal choroidal thickness (r=0.2, P=0.048). CONCLUSION The current study demonstrated that the decreased serum VEGFR-2 level may be considered in the development of AMD. Adropin does not seem to play a role in the pathogenesis of AMD.
Collapse
Affiliation(s)
- Nurgül Örnek
- Department of Ophthalmology, School of Medicine, Kirikkale University, Kirikkale 71450, Turkey
| | - Kemal Örnek
- Department of Ophthalmology, School of Medicine, Kirikkale University, Kirikkale 71450, Turkey
| | - Süleyman Aydin
- Department of Biochemistry, School of Medicine, Firat University, Elazig 23300, Turkey
| | - Musa Yilmaz
- Department of Biochemistry, School of Medicine, Firat University, Elazig 23300, Turkey
| | - Yaşar Ölmez
- Department of Ophthalmology, School of Medicine, Kirikkale University, Kirikkale 71450, Turkey
| |
Collapse
|
196
|
Lieb V, Rink M, Sikic D, Keck B. [Side effect management of tyrosine kinase inhibitors in urology : Gastrointestinal side effects]. Urologe A 2016; 55:805-12. [PMID: 27146873 DOI: 10.1007/s00120-016-0090-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
For approximately one decade, tyrosinkinase inhibitors (TKIs, smart drugs) have dramatically changed and improved the treatment of patients suffering from metastasized renal cell carcinoma. However, the different drugs have substantial side effects. Especially gastrointestinal symptoms may be problematic for patients. These side effects represent a challenge for the physician. On the one hand, dosage modifications and treatment interruption should be avoided to minimize the risk for progression. On the other hand, only mild side effects are tolerable for the patient. Based on a literature review, a clear overview of the incidence of possible side effects for the drugs axitinib, cabozantinib, pazopanib, sorafenib, and sunitinib is provided. Furthermore, we give a practical guide on how to prevent and treat the different gastrointestinal side effects. Finally, it is pointed out when dosage modifications or interruption of treatment are necessary and how to expeditiously re-escalate the treatment after mitigation of side effects.
Collapse
Affiliation(s)
- V Lieb
- Urologische Universitätsklinik Erlangen, Universitätsklinikum Erlangen, Rathsberger Str. 57, 91054, Erlangen, Deutschland
| | - M Rink
- Klinik und Poliklinik für Urologie, Universitätsklinikum Hamburg Eppendorf, Hamburg, Deutschland
| | - D Sikic
- Urologische Universitätsklinik Erlangen, Universitätsklinikum Erlangen, Rathsberger Str. 57, 91054, Erlangen, Deutschland
| | - B Keck
- Urologische Universitätsklinik Erlangen, Universitätsklinikum Erlangen, Rathsberger Str. 57, 91054, Erlangen, Deutschland.
| |
Collapse
|
197
|
Jansson L, Barbu A, Bodin B, Drott CJ, Espes D, Gao X, Grapensparr L, Källskog Ö, Lau J, Liljebäck H, Palm F, Quach M, Sandberg M, Strömberg V, Ullsten S, Carlsson PO. Pancreatic islet blood flow and its measurement. Ups J Med Sci 2016; 121:81-95. [PMID: 27124642 PMCID: PMC4900068 DOI: 10.3109/03009734.2016.1164769] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Pancreatic islets are richly vascularized, and islet blood vessels are uniquely adapted to maintain and support the internal milieu of the islets favoring normal endocrine function. Islet blood flow is normally very high compared with that to the exocrine pancreas and is autonomously regulated through complex interactions between the nervous system, metabolites from insulin secreting β-cells, endothelium-derived mediators, and hormones. The islet blood flow is normally coupled to the needs for insulin release and is usually disturbed during glucose intolerance and overt diabetes. The present review provides a brief background on islet vascular function and especially focuses on available techniques to measure islet blood perfusion. The gold standard for islet blood flow measurements in experimental animals is the microsphere technique, and its advantages and disadvantages will be discussed. In humans there are still no methods to measure islet blood flow selectively, but new developments in radiological techniques hold great hopes for the future.
Collapse
Affiliation(s)
- Leif Jansson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- CONTACT Leif Jansson, Department of Medical Cell Biology, Biomedical Centre, Box 571, Husargatan 3, SE-75123 Uppsala, Sweden
| | - Andreea Barbu
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Birgitta Bodin
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Carl Johan Drott
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Daniel Espes
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Xiang Gao
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Liza Grapensparr
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Örjan Källskog
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Joey Lau
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Hanna Liljebäck
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Fredrik Palm
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - My Quach
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Monica Sandberg
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | | | - Sara Ullsten
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Per-Ola Carlsson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
198
|
Hegan PS, Lanahan AA, Simons M, Mooseker MS. Myosin VI and cardiomyopathy: Left ventricular hypertrophy, fibrosis, and both cardiac and pulmonary vascular endothelial cell defects in the Snell's waltzer mouse. Cytoskeleton (Hoboken) 2016; 72:373-87. [PMID: 26265212 DOI: 10.1002/cm.21236] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 07/30/2015] [Accepted: 08/07/2015] [Indexed: 12/17/2022]
Abstract
In mice and humans, loss of myosin VI (Myo6) function results in deafness, and certain Myo6 mutations also result in cardiomyopathies in humans. The current studies have utilized the Snell's waltzer (sv) mouse (a functional null mutation for Myo6) to determine if this mouse also exhibits cardiac defects and thus used to determine the cellular and molecular basis for Myo6-associated heart disease. Myo6 is expressed in mouse heart where it is predominantly expressed in vascular endothelial cells (VECs) based on co-localization with the VEC cell marker CD31. Sv/sv heart mass is significantly greater than that of sv/+ littermates, a result of left ventricle hypertrophy. The left ventricle of the sv/sv exhibits extensive fibrosis, both interstitial and perivascular, based on histologic staining, and immunolocalization of several markers for fibrosis including fibronectin, collagen IV, and the fibroblast marker vimentin. Myo6 is also expressed in lung VECs but not in VECs of intestine, kidney, or liver. Sv/sv lungs exhibit increased periaveolar fibrosis and enlarged air sacs. Electron microscopy of sv/sv cardiac and lung VECs revealed abnormal ultrastructure, including luminal protrusions and increased numbers of cytoplasmic vesicles. Previous studies have shown that loss of function of either Myo6 or its adaptor binding partner synectin/GIPC results in impaired arterial development due to defects in VEGF signaling. However, examination of synectin/GIPC-/- heart revealed no fibrosis or significantly altered VEC ultrastructure, suggesting that the cardiac and lung defects observed in the sv/sv mouse are not due to Myo6 function in arterial development.
Collapse
Affiliation(s)
- Peter S Hegan
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut
| | - Anthony A Lanahan
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Michael Simons
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut.,Department of Cell Biology, Yale School of Medicine, New Haven, Connecticut
| | - Mark S Mooseker
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut.,Department of Cell Biology, Yale School of Medicine, New Haven, Connecticut.,Department of Pathology, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
199
|
The effects of VEGF-A-inhibitors aflibercept and ranibizumab on the ciliary body and iris of monkeys. Graefes Arch Clin Exp Ophthalmol 2016; 254:1117-25. [PMID: 27106625 DOI: 10.1007/s00417-016-3344-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 03/28/2016] [Accepted: 04/04/2016] [Indexed: 10/21/2022] Open
Abstract
PURPOSE To investigate the effects of intravitreal ranibizumab (Lucentis®) and aflibercept (Eylea®) on the ciliary body and the iris of 12 cynomolgus monkeys with regard to the fenestrations of their blood vessels. MATERIALS AND METHODS Structural changes in the ciliary body and in the iris were investigated with light, fluorescent, and transmission electron microscopy (TEM). The latter was used to specifically quantify fenestrations of the endothelium of blood vessels after treatment with aflibercept and ranibizumab. Each of the two ciliary bodies treated with aflibercept and the two treated with ranibizumab and their controls were examined after 1 and 7 days respectively. Ophthalmological investigations including funduscopy and intraocular pressure measurements were also applied. RESULTS Ophthalmological investigations did not reveal any changes within the groups. Both drugs reduced the VEGF concentration in the ciliary body pigmented epithelium. The structure of the ciliary body was not influenced, while the posterior pigmented epithelium of the iris showed vacuoles after aflibercept treatment. Ranibizumab was mainly concentrated on the surface layer of the ciliary epithelium, in the blood vessel walls and the lumen of some of the blood vessels, and in the cells of the epithelium of the ciliary body. Aflibercept was more concentrated in the stroma and not in the cells of the epithelium, but as with ranibizumab, also in the blood vessel walls and some of their lumina, and again on the surface layer of the epithelium. Both aflibercept-and ranibizumab-treated eyes showed a decreased number of fenestrations of the capillaries in the ciliary body compared to the untreated controls. On day 1 and day 7, aflibercept had fewer fenestrations than the ranibizumab samples of the same day. CONCLUSIONS Both aflibercept and ranibizumab were found to reach the blood vessel walls of the ciliary body, and effectively reduced their fenestrations. Aflibercept might eliminate VEGF to a greater extent, possibly due to a higher elimination of fenestrations in a shorter time. Moreover, the vacuoles found in the iris need further research, in order to evaluate whether they carry a possible pathological potential.
Collapse
|
200
|
Peng P, Wang C, Tao S, Sun C. The Deficit Profiles of Chinese Children with Reading Difficulties: a Meta-analysis. EDUCATIONAL PSYCHOLOGY REVIEW 2016. [DOI: 10.1007/s10648-016-9366-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|