1
|
Pączek S, Zajkowska M, Mroczko B. Pigment Epithelial-Derived Factor in Pancreatic and Liver Cancers-From Inflammation to Cancer. Biomedicines 2024; 12:2260. [PMID: 39457573 PMCID: PMC11504982 DOI: 10.3390/biomedicines12102260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/28/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024] Open
Abstract
Gastrointestinal (GI) cancers are among the leading causes of mortality worldwide. Despite the emergence of new possibilities that offer hope regarding the successful treatment of these cancers, they still represent a significant global health burden. These cancers can arise from various cell types within the gastrointestinal tract and may exhibit different characteristics, behaviors, and treatment approaches. Both the prognosis and the outcomes of GI treatment remain problematic because these tumors are primarily diagnosed in advanced clinical stages. Current biomarkers exhibit limited sensitivity and specificity. Therefore, when developing strategies for the diagnosis and treatment of GI cancers, it is of fundamental importance to discover new biomarkers capable of addressing the challenges of early-stage diagnosis and the presence of lymph node metastases. Pigment epithelial-derived factor (PEDF) has garnered interest due to its inhibitory effects on the migration and proliferation of cancer cells. This protein has been suggested to be involved in various inflammation-related diseases, including cancer, through various mechanisms. It was also observed that reducing the level of PEDF is sufficient to trigger an inflammatory response. This suggests that PEDF is an endogenous anti-inflammatory factor. Overall, PEDF is a versatile protein with diverse biological functions that span across different tissues and organ systems. Its multifaceted activities make it an intriguing target for therapeutic interventions in various diseases, including cancer, neurodegeneration, and metabolic disorders. This review, for the first time, summarizes the role of PEDF in the pathogenesis of selected GI cancers and its potential utility in early diagnosis, prognosis, and therapeutic strategies for this malignancy.
Collapse
Affiliation(s)
- Sara Pączek
- Department of Biochemical Diagnostics, University Hospital in Białystok, 15-269 Białystok, Poland; (S.P.); (B.M.)
| | - Monika Zajkowska
- Department of Biochemical Diagnostics, University Hospital in Białystok, 15-269 Białystok, Poland; (S.P.); (B.M.)
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, 15 A, Waszyngtona St., 15-269 Białystok, Poland
| | - Barbara Mroczko
- Department of Biochemical Diagnostics, University Hospital in Białystok, 15-269 Białystok, Poland; (S.P.); (B.M.)
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, 15 A, Waszyngtona St., 15-269 Białystok, Poland
- Department of Biochemical Diagnostics, Medical University of Białystok, 15-089 Białystok, Poland
| |
Collapse
|
2
|
Limbu S, McCloskey KE. An Endothelial Cell Is Not Simply an Endothelial Cell. Stem Cells Dev 2024; 33:517-527. [PMID: 39030822 PMCID: PMC11564855 DOI: 10.1089/scd.2024.0088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/18/2024] [Indexed: 07/22/2024] Open
Abstract
Endothelial cells (ECs) are a multifaceted component of the vascular system with roles in immunity, maintaining tissue fluid balance, and vascular tone. Dysregulation or dysfunction of ECs can have far-reaching implications, leading pathologies ranging from cardiovascular diseases, such as hypertension and atherosclerosis, ischemia, chronic kidney disease, blood-brain barrier integrity, dementia, and tumor metastasis. Recent advancements in regenerative medicine have highlighted the potential of stem cell-derived ECs, particularly from induced pluripotent stem cells, to treat ischemic tissues, as well as models of vascular integrity. This review summarizes what is known in the generation of ECs with an emphasis on tissue-specific ECs and EC subphenotypes important in the development of targeted cell-based therapies for patient treatment.
Collapse
Affiliation(s)
- Shiwani Limbu
- Quantitative and System Biology Graduate Program, University of California, Merced, USA
| | - Kara E. McCloskey
- Quantitative and System Biology Graduate Program, University of California, Merced, USA
- Materials Science and Engineering Department, University of California, Merced, USA
| |
Collapse
|
3
|
Saito J, Dave JM, Lau FD, Greif DM. Presenilin-1 in smooth muscle cells facilitates hypermuscularization in elastin aortopathy. iScience 2024; 27:108636. [PMID: 38226162 PMCID: PMC10788461 DOI: 10.1016/j.isci.2023.108636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 11/16/2023] [Accepted: 12/01/2023] [Indexed: 01/17/2024] Open
Abstract
Smooth muscle cell (SMC) accumulation is central to the pathogenesis of elastin-defective arterial diseases, including supravalvular aortic stenosis (SVAS). We previously demonstrated that elastin insufficiency activates Notch signaling in aortic SMCs. Activation of Notch is catalyzed by the enzyme gamma-secretase, but the role of catalytic subunits presenilin (PSEN)-1 or PSEN-2 in elastin aortopathy is not defined. Genetic approaches reveal that endothelial cell-specific Psen1 deletion does not improve elastin aortopathy whereas the deletion of either Psen1 in SMCs or Psen2 globally attenuates Notch pathway and SMC proliferation, mitigating aortic disease. With SMC-specific Psen1 deletion in elastin nulls, these rescue effects are more robust and in fact, survival is increased. SMC deletion of Psen1 also attenuates hypermuscularization in newborns heterozygous for the elastin null gene, which genetically mimics SVAS. Similarly, the pharmacological inhibition of PSEN-1 mitigates SMC accumulation in elastin aortopathy. These findings put forth SMC PSEN-1 as a potential therapeutic target in SVAS.
Collapse
Affiliation(s)
- Junichi Saito
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
- Department of Genetics, Yale University, New Haven, CT 06511, USA
- Stem Cell Center, Yale University, New Haven, CT 06511, USA
| | - Jui M. Dave
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
- Department of Genetics, Yale University, New Haven, CT 06511, USA
- Stem Cell Center, Yale University, New Haven, CT 06511, USA
| | - Freddy Duarte Lau
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
- Department of Genetics, Yale University, New Haven, CT 06511, USA
| | - Daniel M. Greif
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
- Department of Genetics, Yale University, New Haven, CT 06511, USA
- Stem Cell Center, Yale University, New Haven, CT 06511, USA
| |
Collapse
|
4
|
Deng C, Cai Q, Zhang J, Chang K, Peng T, Liu X, Cao F, Yan X, Cheng J, Wang X, Tan Y, Hua Q. Generation and Characterization of a Novel Knockin Mouse Model Expressing PSEN1 D385A: Implications for Investigating Herbal Drug Effects in γ-Secretase Activity. J Alzheimers Dis 2024; 100:825-841. [PMID: 38905042 DOI: 10.3233/jad-231148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/23/2024]
Abstract
Background Presenilin (PSEN, PS) is essential for γ-secretase function, and mutations can disrupt amyloid-β (Aβ) production in familial Alzheimer's disease. Targeting γ-secretase is complex due to its broad involvement in physiological processes. Objective Our aim was to create a novel knockin (KI) mouse model expressing PSEN1 D385A mutation and investigate the efficacy of a Geniposide and Ginsenoside Rg1 combination (NeuroProtect modified formula, NP-2) in restoring γ-secretase activity. Methods Using gene manipulation, we established the PS1 D385A KI mouse model and confirmed the mutation, mRNA, and protein levels using Southern blotting, northern blotting, and western blotting, respectively. In vitro γ-secretase assay was conducted to measure γ-secretase activity, while histological analyses examined neurogenesis effects. NP-2 administration evaluated its impact on γ-secretase activity. Results The PS1 D385A KI homozygotes displayed severe cerebral hemorrhage, postnatal lethality, developmental disorders, reduced proliferation of neural progenitor cells, and disrupted γ-secretase function. The mutation abolished PS1 protein self-shearing, leading to compromised γ-secretase activity. NP-2 intervention effectively restored γ-secretase activity in the heterozygous mice. Conclusions PS1 D385A mutant disrupted PS1 protein self-cleaving, impairing γ-secretase activity in KI mice. NP-2 restored γ-secretase function, offering potential for novel AD treatment strategies despite the challenges posed by γ-secretase's complex role in physiological processes.
Collapse
Affiliation(s)
- Chengeng Deng
- School of Life Sciences, School of Acupuncture-Moxibustion and Tuina, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qingyuan Cai
- School of Life Sciences, School of Acupuncture-Moxibustion and Tuina, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jiani Zhang
- School of Life Sciences, School of Acupuncture-Moxibustion and Tuina, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Kexin Chang
- School of Life Sciences, School of Acupuncture-Moxibustion and Tuina, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Tiantian Peng
- School of Life Sciences, School of Acupuncture-Moxibustion and Tuina, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoge Liu
- School of Life Sciences, School of Acupuncture-Moxibustion and Tuina, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Feng Cao
- School of Life Sciences, School of Acupuncture-Moxibustion and Tuina, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xinyuan Yan
- School of Life Sciences, School of Acupuncture-Moxibustion and Tuina, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Junshi Cheng
- School of Life Sciences, School of Acupuncture-Moxibustion and Tuina, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xu Wang
- School of Life Sciences, School of Acupuncture-Moxibustion and Tuina, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yan Tan
- School of Life Sciences, School of Acupuncture-Moxibustion and Tuina, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qian Hua
- School of Life Sciences, School of Acupuncture-Moxibustion and Tuina, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
5
|
Wei W, Zhang Y. PSEN1 is associated with colon cancer development via potential influences on PD-L1 nuclear translocation and tumor-immune interactions. Front Immunol 2022; 13:927474. [PMID: 36059511 PMCID: PMC9428321 DOI: 10.3389/fimmu.2022.927474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
Presenilin 1 (PSEN1), as a catalytical core of the γ-secretase complex, plays multiple actions through mediating transmembrane domain shedding of the substrates. Unlike extensive studies performed on investigating the functions of γ-secretase substrates or the effects of γ-secretase inhibitors, our findings uncover a potential action of PSEN1 on PD-L1 alternative truncation and nuclear translocation, broadening our understanding on how the γ-secretase contributes to colon cancer development as well as suggesting a potential strategy to improve the efficacy of PD-1/PD-L1 blockade. Immunohistochemical data showed loss of PD-L1 protein expression in all the primary colon adenocarcioma (COAD) cases in the HPA collection, while PSEN1 was scored to be highly expressed, indicating their converse expression patterns (p<0.001). Meanwhile a strongly positive gene correlation was explored by TIMER2 and GEPIA (p<0.001). Up-regulated PSEN1 expression in COAD might facilitate liberating a C-terminal PD-L1 truncation via proteolytic processing. Then following an established regulatory pathway of PD-L1 nuclear translocation, we found that PSEN1 showed significant correlations with multiple components in HDAC2-mediated deacetylation, clathrin-dependent endocytosis, vimentin-associated nucleocytoplasmic shuttling and importin family-mediated nuclear import. Moreover, connections of PSEN1 to the immune response genes transactivated by nuclear PD-L1 were tested. Additionally, contributions of PSEN1 to the tumor invasiveness (p<0.05) and the tumor infiltrating cell enrichments (p<0.001) were investigated by cBioportal and the ESTIMATE algorithm. Levels of PSEN1 were negatively correlated with infiltrating CD8+ T (p<0.05) and CD4+ T helper (Th) 1 cells (p<0.001), while positively correlated with regulatory T cells (Tregs) (p<0.001) and cancer associated fibroblasts (CAFs) (p<0.001). It also displayed significant associations with diverse immune metagenes characteristic of T cell exhaustion, Tregs and CAFs, indicating possible actions in immune escape. Despite still a preliminary stage of this study, we anticipate to deciphering a novel function of PSEN1, and supporting more researchers toward the elucidations of the mechanisms linking the γ-secretase to cancers, which has yet to be fully addressed.
Collapse
|
6
|
Teplyashina EA, Komleva YK, Lychkovskaya EV, Deikhina AS, Salmina AB. Regulation of neurogenesis and cerebral angiogenesis by cell protein proteolysis products. RUDN JOURNAL OF MEDICINE 2021. [DOI: 10.22363/2313-0245-2021-25-2-114-126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Brain development is a unique process characterized by mechanisms defined as neuroplasticity (synaptogenesis, synapse elimination, neurogenesis, and cerebral angiogenesis). Numerous neurodevelopmental disorders brain damage, and aging are manifested by neurological deficits that are caused by aberrant neuroplasticity. The presence of stem and progenitor cells in neurogenic niches of the brain is responsible for the formation of new neurons capable of integrating into preexisting synaptic assemblies. The determining factors for the cells within the neurogenic niche are the activity of the vascular scaffold and the availability of active regulatory molecules that establish the optimal microenvironment. It has been found that regulated intramembrane proteolysis plays an important role in the control of neurogenesis in brain neurogenic niches. Molecules generated by the activity of specific proteases can stimulate or suppress the activity of neural stem and progenitor cells, their proliferation and differentiation, migration and integration of newly formed neurons into synaptic networks. Local neoangiogenesis supports the processes of neurogenesis in neurogenic niches, which is guaranteed by the multivalent action of peptides formed from transmembrane proteins. Identification of new molecules regulating the neuroplasticity (neurogenesis and angiogenesis). i. e. enzymes, substrates, and products of intramembrane proteolysis, will ensure the development of protocols for detecting the neuroplasticity markers and targets for efficient pharmacological modulation.
Collapse
|
7
|
Sen S, Hallee L, Lam CK. The Potential of Gamma Secretase as a Therapeutic Target for Cardiac Diseases. J Pers Med 2021; 11:jpm11121294. [PMID: 34945766 PMCID: PMC8703931 DOI: 10.3390/jpm11121294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/24/2021] [Accepted: 12/01/2021] [Indexed: 11/16/2022] Open
Abstract
Heart diseases are some of the most common and pressing threats to human health worldwide. The American Heart Association and the National Institute of Health jointly work to annually update data on cardiac diseases. In 2018, 126.9 million Americans were reported as having some form of cardiac disorder, with an estimated direct and indirect total cost of USD 363.4 billion. This necessitates developing therapeutic interventions for heart diseases to improve human life expectancy and economic relief. In this review, we look into gamma-secretase as a potential therapeutic target for cardiac diseases. Gamma-secretase, an aspartyl protease enzyme, is responsible for the cleavage and activation of a number of substrates that are relevant to normal cardiac development and function as found in mutation studies. Some of these substrates are involved in downstream signaling processes and crosstalk with pathways relevant to heart diseases. Most of the substrates and signaling events we explored were found to be potentially beneficial to maintain cardiac function in diseased conditions. This review presents an updated overview of the current knowledge on gamma-secretase processing of cardiac-relevant substrates and seeks to understand if the modulation of gamma-secretase activity would be beneficial to combat cardiac diseases.
Collapse
Affiliation(s)
- Sujoita Sen
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA;
| | - Logan Hallee
- Department of Mathematical Sciences, University of Delaware, Newark, DE 19716, USA;
| | - Chi Keung Lam
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA;
- Correspondence: ; Tel.: +1-302-831-3165
| |
Collapse
|
8
|
Farooq M, Abutaha N, Mahboob S, Baabbad A, Almoutiri ND, Wadaan MAAM. Investigating the antiangiogenic potential of Rumex vesicarius (humeidh), anticancer activity in cancer cell lines and assessment of developmental toxicity in zebrafish embryos. Saudi J Biol Sci 2020; 27:611-622. [PMID: 32210679 PMCID: PMC6997907 DOI: 10.1016/j.sjbs.2019.11.042] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 11/26/2019] [Accepted: 11/28/2019] [Indexed: 12/29/2022] Open
Abstract
Recent trends in anticancer therapy is to use therapeutic agents which not only kill the cancer cell, but are less toxic to surrounding normal cells/tissue. One approach is to cut the nutrient supply to growing tumor cells, by blocking the formation of new blood vessels around the tumor. As the phytochemicals and botanical crude extracts have proven their efficacy as natural antiangiogenic agents with minimum toxicities, there is need to explore varieties of medicinal plants for novel antiangiogenic compounds. Rumex vesicarius L. (Humeidh), is an annual herbal plant with proven medicinal values. The antiangiogenic potential, and developmental toxicity of humeidh in experimental animal models has never been studied before. The crude extracts were prepared from the roots, stems, leaves and flowers of Rumex vesicarius L. in methanol, chloroform, ethyl acetate and n-hexane. The developmental toxicity screening in zebrafish embryos, has revealed that Rumex vesicarius was not toxic to zebrafish embryos. The chloroform stem extract showed significant level of antiangiogenic activity in zebrafish angiogenic assay on a dose dependent manner. Thirty five (35) bioactive compounds were identified by gas chromatography mass spectrophotometry (GC–MS) analysis in the stem extract of Rumex vesicarius. Propanoic acid, 2-[(trimethylsilyl)oxy]-, trimethylsilyl ester, Butane, 1,2,3-tris(trimethylsiloxy), and Butanedioic acid, bis(trimethylsilyl) ester were identified as major compound present in the stem of R. vasicarius. The anticancer activity of roots, stem, leaves and flowers crude extract was evaluated in human breast cancer (MCF7), human colon carcinoma (Lovo, and Caco-2), human hepatocellular carcinoma (HepG2) cell lines. Most of the crude extracts did not show significant level of cytotoxicity in tested cancer cells line, except, chloroform extract of stem which exhibited strong anticancer activity in all tested cancer cells with IC50 values in micro molar range. Based on these results, it is recommended that formulation prepared from R. vesicarius can further be tested in clinical trials in order to explore its therapeutic potential as an effective and safe natural anticancer product.
Collapse
Affiliation(s)
- Muhammad Farooq
- College of Science, Department of Zoology, King Saud University, 11451 Riyadh, Saudi Arabia
| | - Nael Abutaha
- College of Science, Department of Zoology, King Saud University, 11451 Riyadh, Saudi Arabia
| | - Shahid Mahboob
- Department of Zoology, College of Science, King Saud University, 11451 Riyadh, Saudi Arabia
| | - Almohannad Baabbad
- College of Science, Department of Zoology, King Saud University, 11451 Riyadh, Saudi Arabia
| | - Nawaf D Almoutiri
- College of Science, Department of Zoology, King Saud University, 11451 Riyadh, Saudi Arabia
| | | |
Collapse
|
9
|
Logan SM, Storey KB. Angiogenic signaling in the lungs of a metabolically suppressed hibernating mammal ( Ictidomys tridecemlineatus). PeerJ 2019; 7:e8116. [PMID: 31763078 PMCID: PMC6870509 DOI: 10.7717/peerj.8116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 10/29/2019] [Indexed: 01/18/2023] Open
Abstract
To conserve energy in times of limited resource availability, particularly during cold winters, hibernators suppress even the most basic of physiologic processes. Breathing rates decrease from 40 breaths/minute to less than 1 breath/min as they decrease body temperature from 37 °C to ambient. Nevertheless, after months of hibernation, these incredible mammals emerge from torpor unscathed. This study was conducted to better understand the protective and possibly anti-inflammatory adaptations that hibernator lungs may use to prevent damage associated with entering and emerging from natural torpor. We postulated that the differential protein expression of soluble protein receptors (decoy receptors that sequester soluble ligands to inhibit signal transduction) would help identify inhibited inflammatory signaling pathways in metabolically suppressed lungs. Instead, the only two soluble receptors that responded to torpor were sVEGFR1 and sVEGFR2, two receptors whose full-length forms are bound by VEGF-A to regulate endothelial cell function and angiogenesis. Decreased sVEGFR1/2 correlated with increased total VEGFR2 protein levels. Maintained or increased levels of key γ-secretase subunits suggested that decreased sVEGFR1/2 protein levels were not due to decreased levels of intramembrane cleavage complex subunits. VEGF-A protein levels did not change, suggesting that hibernators may regulate VEGFR1/2 signaling at the level of the receptor instead of increasing relative ligand abundance. A panel of angiogenic factors used to identify biomarkers of angiogenesis showed a decrease in FGF-1 and an increase in BMP-9. Torpid lungs may use VEGF and BMP-9 signaling to balance angiogenesis and vascular stability, possibly through the activation of SMAD signaling for adaptive tissue remodeling.
Collapse
Affiliation(s)
- Samantha M. Logan
- Institute of Biochemistry, Departments of Biology and Chemistry, Carleton University, Ottawa, ON, Canada
| | - Kenneth B. Storey
- Institute of Biochemistry, Departments of Biology and Chemistry, Carleton University, Ottawa, ON, Canada
| |
Collapse
|
10
|
Affiliation(s)
- Mary G Sorci-Thomas
- From the Departments of Medicine, Section on Endocrinology (M.G.S.-T.) and Pharmacology and Toxicology (M.G.S.-T., M.J.T.), Medical College of Wisconsin, Milwaukee.
| | - Michael J Thomas
- From the Departments of Medicine, Section on Endocrinology (M.G.S.-T.) and Pharmacology and Toxicology (M.G.S.-T., M.J.T.), Medical College of Wisconsin, Milwaukee
| |
Collapse
|
11
|
Warren NA, Voloudakis G, Yoon Y, Robakis NK, Georgakopoulos A. The product of the γ-secretase processing of ephrinB2 regulates VE-cadherin complexes and angiogenesis. Cell Mol Life Sci 2018; 75:2813-2826. [PMID: 29428965 PMCID: PMC6023733 DOI: 10.1007/s00018-018-2762-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 11/25/2017] [Accepted: 01/25/2018] [Indexed: 01/01/2023]
Abstract
Presenilin-1 (PS1) gene encodes the catalytic component of γ-secretase, which proteolytically processes several type I transmembrane proteins. We here present evidence that the cytosolic peptide efnB2/CTF2 produced by the PS1/γ-secretase cleavage of efnB2 ligand promotes EphB4 receptor-dependent angiogenesis in vitro. EfnB2/CTF2 increases endothelial cell sprouting and tube formation, stimulates the formation of angiogenic complexes that include VE-cadherin, Raf-1 and Rok-α, and increases MLC2 phosphorylation. These functions are mediated by the PDZ-binding domain of efnB2. Acute downregulation of PS1 or inhibition of γ-secretase inhibits the angiogenic functions of EphB4 while absence of PS1 decreases the VE-cadherin angiogenic complexes of mouse brain. Our data reveal a mechanism by which PS1/γ-secretase regulates efnB2/EphB4 mediated angiogenesis.
Collapse
Affiliation(s)
- Noel A Warren
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Georgios Voloudakis
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yonejung Yoon
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nikolaos K Robakis
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anastasios Georgakopoulos
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
12
|
Gupta V, Gupta VB, Chitranshi N, Gangoda S, Vander Wall R, Abbasi M, Golzan M, Dheer Y, Shah T, Avolio A, Chung R, Martins R, Graham S. One protein, multiple pathologies: multifaceted involvement of amyloid β in neurodegenerative disorders of the brain and retina. Cell Mol Life Sci 2016; 73:4279-4297. [PMID: 27333888 PMCID: PMC11108534 DOI: 10.1007/s00018-016-2295-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 06/14/2016] [Accepted: 06/15/2016] [Indexed: 01/18/2023]
Abstract
Accumulation of amyloid β (Aβ) and its aggregates in the ageing central nervous system is regarded synonymous to Alzheimer's disease (AD) pathology. Despite unquestionable advances in mechanistic and diagnostic aspects of the disease understanding, the primary cause of Aβ accumulation as well as its in vivo roles remains elusive; nonetheless, the majority of the efforts to address pathological mechanisms for therapeutic development are focused towards moderating Aβ accumulation in the brain. More recently, Aβ deposition has been identified in the eye and is linked with distinct age-related diseases including age-related macular degeneration, glaucoma as well as AD. Awareness of the Aβ accumulation in these markedly different degenerative disorders has led to an increasing body of work exploring overlapping mechanisms, a prospective biomarker role for Aβ and the potential to use retina as a model for brain related neurodegenerative disorders. Here, we present an integrated view of current understanding of the retinal Aβ deposition discussing the accumulation mechanisms, anticipated impacts and outlining ameliorative approaches that can be extrapolated to the retina for potential therapeutic benefits. Further longitudinal investigations in humans and animal models will determine retinal Aβ association as a potential pathognomonic, diagnostic or prognostic biomarker.
Collapse
Affiliation(s)
- Vivek Gupta
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Veer B Gupta
- School of Medical Sciences, Edith Cowan University, Perth, Australia.
| | - Nitin Chitranshi
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Sumudu Gangoda
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Roshana Vander Wall
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Mojdeh Abbasi
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Mojtaba Golzan
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Yogita Dheer
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Tejal Shah
- School of Medical Sciences, Edith Cowan University, Perth, Australia
| | - Alberto Avolio
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Roger Chung
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Ralph Martins
- School of Medical Sciences, Edith Cowan University, Perth, Australia
| | - Stuart Graham
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
- Save Sight Institute, Sydney University, Sydney, Australia
| |
Collapse
|
13
|
Li P, Lin X, Zhang JR, Li Y, Lu J, Huang FC, Zheng CH, Xie JW, Wang JB, Huang CM. The expression of presenilin 1 enhances carcinogenesis and metastasis in gastric cancer. Oncotarget 2016; 7:10650-62. [PMID: 26872378 PMCID: PMC4891148 DOI: 10.18632/oncotarget.7298] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 01/19/2016] [Indexed: 12/12/2022] Open
Abstract
Presenilin 1 (PS-1, encoded by PSEN1) is a part of the gamma- (γ-) secretase complex. Mutations in PSEN1 cause the majority of cases of familial Alzheimer's disease (FAD). Although in recent years PS-1 has been implicated as a tumor enhancer in various cancers, nothing is known regarding its role in gastric cancer (GC). In the present study, we investigate the role and clinical significance of PS-1 in GC. We observed that PS-1 was significantly upregulated and amplified in GC tissues and cell lines, and its aberrant expression was positively correlated with lymph node metastasis and with poor overall survival. Furthermore, PS-1 promoted tumor invasion and metastasis of GC both in vitro and vivo without affecting the proliferation of GC cells (MGC-803 and MKN-45). The results of treatment with the γ-secretase inhibitor DAPT were consistent with the outcomes of PS-1 silencing. PS-1/γ-secretase cleaves E-cadherin and releases its bound protein partner, β-catenin, from the actin cytoskeleton, thereby allowing it to translocate into the nucleus and to activate the TCF/LEF-1 transcriptional activator, which may promote GC invasion and metastasis.In conclusion, PS-1 promotes invasion and metastasis in GC and may represent a novel prognostic biomarker and potential therapeutic target for GC treatment.
Collapse
Affiliation(s)
- Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| | - Xi Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| | - Jun-Rong Zhang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| | - Yun Li
- Key Laboratory of the Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People's Republic of China
| | - Jun Lu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| | - Fei-Chao Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| | - Jia-Bin Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| |
Collapse
|
14
|
Mahadevaiah S, Robinson KG, Kharkar PM, Kiick KL, Akins RE. Decreasing matrix modulus of PEG hydrogels induces a vascular phenotype in human cord blood stem cells. Biomaterials 2015; 62:24-34. [PMID: 26016692 DOI: 10.1016/j.biomaterials.2015.05.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 05/04/2015] [Accepted: 05/14/2015] [Indexed: 01/12/2023]
Abstract
Adult and congenital cardiovascular diseases are significant health problems that are often managed using surgery. Bypass grafting is a principal therapy, but grafts fail at high rates due to hyperplasia, fibrosis, and atherosclerosis. Biocompatible, cellularized materials that attenuate these complications and encourage healthy microvascularization could reduce graft failure, but an improved understanding of biomaterial effects on human stem cells is needed to reach clinical utility. Our group investigates stem-cell-loaded biomaterials for placement along the adventitia of at-risk vessels and grafts. Here, the effects of substrate modulus on human CD34+ stem cells from umbilical cord blood were evaluated. Cells were isolated by immunomagnetic separation and encapsulated in 3, 4, and 6 weight% PEG hydrogels containing 0.032% gelatin and 0.0044% fibronectin. Gels reached moduli of 0.34, 4.5, and 9.1 kPa. Cell viability approached 100%. Cell morphologies appeared similar across gels, but proliferation was significantly lower in 6 wt% gels. Expression profiling using stem cell signaling arrays indicated enhanced self-renewal and differentiation into vascular endothelium among cells in the lower weight percent gels. Thus, modulus was associated with cell proliferation and function. Gels with moduli in the low kilopascal range may be useful in stimulating cell engraftment and microvascularization of graft adventitia.
Collapse
Affiliation(s)
- Shruthi Mahadevaiah
- Nemours - Alfred I. duPont Hospital for Children, Department of Biomedical Research, 1600 Rockland Road, Wilmington, DE 19803, United States; Nemours - Alfred I. duPont Hospital for Children, Critical Care Department, 1600 Rockland Road, Wilmington, DE 19803, United States
| | - Karyn G Robinson
- Nemours - Alfred I. duPont Hospital for Children, Department of Biomedical Research, 1600 Rockland Road, Wilmington, DE 19803, United States
| | - Prathamesh M Kharkar
- Department of Materials Science and Engineering, University of Delaware, 201 Du Pont Hall, Newark, DE 19716, United States
| | - Kristi L Kiick
- Department of Materials Science and Engineering, University of Delaware, 201 Du Pont Hall, Newark, DE 19716, United States
| | - Robert E Akins
- Nemours - Alfred I. duPont Hospital for Children, Department of Biomedical Research, 1600 Rockland Road, Wilmington, DE 19803, United States.
| |
Collapse
|
15
|
Reichhart N, Keckeis S, Fried F, Fels G, Strauss O. Regulation of surface expression of TRPV2 channels in the retinal pigment epithelium. Graefes Arch Clin Exp Ophthalmol 2015; 253:865-74. [DOI: 10.1007/s00417-014-2917-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 12/23/2014] [Accepted: 12/23/2014] [Indexed: 12/31/2022] Open
|
16
|
Peng Y, Li Z, Yang P, Newton IP, Ren H, Zhang L, Wu H, Li Z. Direct contacts with colon cancer cells regulate the differentiation of bone marrow mesenchymal stem cells into tumor associated fibroblasts. Biochem Biophys Res Commun 2014; 451:68-73. [DOI: 10.1016/j.bbrc.2014.07.074] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 07/15/2014] [Indexed: 12/13/2022]
|
17
|
Niedowicz DM, Reeves VL, Platt TL, Kohler K, Beckett TL, Powell DK, Lee TL, Sexton TR, Song ES, Brewer LD, Latimer CS, Kraner SD, Larson KL, Ozcan S, Norris CM, Hersh LB, Porter NM, Wilcock DM, Murphy MP. Obesity and diabetes cause cognitive dysfunction in the absence of accelerated β-amyloid deposition in a novel murine model of mixed or vascular dementia. Acta Neuropathol Commun 2014; 2:64. [PMID: 24916066 PMCID: PMC4229778 DOI: 10.1186/2051-5960-2-64] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 06/04/2014] [Indexed: 01/18/2023] Open
Abstract
Mid-life obesity and type 2 diabetes mellitus (T2DM) confer a modest, increased risk for Alzheimer's disease (AD), though the underlying mechanisms are unknown. We have created a novel mouse model that recapitulates features of T2DM and AD by crossing morbidly obese and diabetic db/db mice with APPΔNL/ΔNLx PS1P264L/P264L knock-in mice. These mice (db/AD) retain many features of the parental lines (e.g. extreme obesity, diabetes, and parenchymal deposition of β-amyloid (Aβ)). The combination of the two diseases led to additional pathologies-perhaps most striking of which was the presence of severe cerebrovascular pathology, including aneurysms and small strokes. Cortical Aβ deposition was not significantly increased in the diabetic mice, though overall expression of presenilin was elevated. Surprisingly, Aβ was not deposited in the vasculature or removed to the plasma, and there was no stimulation of activity or expression of major Aβ-clearing enzymes (neprilysin, insulin degrading enzyme, or endothelin-converting enzyme). The db/AD mice displayed marked cognitive impairment in the Morris Water Maze, compared to either db/db or APPΔNLx PS1P264L mice. We conclude that the diabetes and/or obesity in these mice leads to a destabilization of the vasculature, leading to strokes and that this, in turn, leads to a profound cognitive impairment and that this is unlikely to be directly dependent on Aβ deposition. This model of mixed or vascular dementia provides an exciting new avenue of research into the mechanisms underlying the obesity-related risk for age-related dementia, and will provide a useful tool for the future development of therapeutics.
Collapse
|
18
|
Loss of presenilin 2 is associated with increased iPLA2 activity and lung tumor development. Oncogene 2014; 33:5193-200. [PMID: 24858037 PMCID: PMC4287650 DOI: 10.1038/onc.2014.128] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 04/11/2014] [Accepted: 04/14/2014] [Indexed: 12/17/2022]
Abstract
Presenilins are the enzymatic components of γ-secretase complex that cleaves amyloid precursor protein, Notch and β-catenin, which has critical roles in the development of Alzheimer's disease and cancer cell growth. Therefore, in the present study, we studied the effects and mechanisms of PS2 knockout on lung cancer development and possible mechanisms as a key regulator of lung tumor development. We compared carcinogen-induced tumor growth between PS2 knockout mice and wild-type mice. PS2 knockout mice showed increased urethane (1 mg/g)-induced lung tumor incidence when compared with that of wild-type mice with decreased activity of γ-secretase in the lung tumor tissues. Consequently, iPLA2 activities in lung tumor tissues of PS2 knockout mice were much higher than in tumor tissues of wild-type mice. Furthermore, knockdown of PS2 using PS2 siRNA decreased γ-secretase activity with increased iPLA2 activity in the lung cancer cells (A549 and NCI-H460), leading to increased lung cancer cell growth. PS2 knockout mice and PS2 knockdown lung cancer cells showed increased DNA-binding activities of nuclear factor kappa-beta, signal transducer and activator of transcription 3 (STAT3) and AP-1 which are critical transcriptional factors of iPLA2 than those of PS2 wild-type mice and control lung cancer cells. Taken together, these results suggest that the loss of PS2 could have a critical role in lung tumor development through the upregulation of iPLA2 activity by reducing γ-secretase.
Collapse
|
19
|
Hu J, Popp R, Frömel T, Ehling M, Awwad K, Adams RH, Hammes HP, Fleming I. Müller glia cells regulate Notch signaling and retinal angiogenesis via the generation of 19,20-dihydroxydocosapentaenoic acid. ACTA ACUST UNITED AC 2014; 211:281-95. [PMID: 24446488 PMCID: PMC3920554 DOI: 10.1084/jem.20131494] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Cytochrome P450 (CYP) epoxygenases generate bioactive lipid epoxides which can be further metabolized to supposedly less active diols by the soluble epoxide hydrolase (sEH). As the role of epoxides and diols in angiogenesis is unclear, we compared retinal vasculature development in wild-type and sEH(-/-) mice. Deletion of the sEH significantly delayed angiogenesis, tip cell, and filopodia formation, a phenomenon associated with activation of the Notch signaling pathway. In the retina, sEH was localized in Müller glia cells, and Müller cell-specific sEH deletion reproduced the sEH(-/-) retinal phenotype. Lipid profiling revealed that sEH deletion decreased retinal and Müller cell levels of 19,20-dihydroxydocosapentaenoic acid (DHDP), a diol of docosahexenoic acid (DHA). 19,20-DHDP suppressed endothelial Notch signaling in vitro via inhibition of the γ-secretase and the redistribution of presenilin 1 from lipid rafts. Moreover, 19,20-DHDP, but not the parent epoxide, was able to rescue the defective angiogenesis in sEH(-/-) mice as well as in animals lacking the Fbxw7 ubiquitin ligase, which demonstrate strong basal activity of the Notch signaling cascade. These studies demonstrate that retinal angiogenesis is regulated by a novel form of neuroretina-vascular interaction involving the sEH-dependent generation of a diol of DHA in Müller cells.
Collapse
Affiliation(s)
- Jiong Hu
- Institute for Vascular Signaling, Centre for Molecular Medicine, Johann Wolfgang Goethe University and DZHK (German Centre for Cardiovascular Research) partner site Rhine-Main, 60590 Frankfurt, Germany
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Podoplanin is a substrate of presenilin-1/γ-secretase. Int J Biochem Cell Biol 2014; 46:68-75. [DOI: 10.1016/j.biocel.2013.11.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 10/17/2013] [Accepted: 11/05/2013] [Indexed: 11/24/2022]
|
21
|
Jurisch-Yaksi N, Sannerud R, Annaert W. A fast growing spectrum of biological functions of γ-secretase in development and disease. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1828:2815-27. [PMID: 24099003 DOI: 10.1016/j.bbamem.2013.04.016] [Citation(s) in RCA: 110] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 04/03/2013] [Accepted: 04/11/2013] [Indexed: 12/17/2022]
Abstract
γ-secretase, which assembles as a tetrameric complex, is an aspartyl protease that proteolytically cleaves substrate proteins within their membrane-spanning domain; a process also known as regulated intramembrane proteolysis (RIP). RIP regulates signaling pathways by abrogating or releasing signaling molecules. Since the discovery, already >15 years ago, of its catalytic component, presenilin, and even much earlier with the identification of amyloid precursor protein as its first substrate, γ-secretase has been commonly associated with Alzheimer's disease. However, starting with Notch and thereafter a continuously increasing number of novel substrates, γ-secretase is becoming linked to an equally broader range of biological processes. This review presents an updated overview of the current knowledge on the diverse molecular mechanisms and signaling pathways controlled by γ-secretase, with a focus on organ development, homeostasis and dysfunction. This article is part of a Special Issue entitled: Intramembrane Proteases.
Collapse
Affiliation(s)
- Nathalie Jurisch-Yaksi
- Laboratory for Membrane Trafficking, VIB-Center for the Biology of Disease & Department for Human Genetics (KU Leuven), Leuven, Belgium
| | | | | |
Collapse
|
22
|
Peltonen HM, Haapasalo A, Hiltunen M, Kataja V, Kosma VM, Mannermaa A. Γ-secretase components as predictors of breast cancer outcome. PLoS One 2013; 8:e79249. [PMID: 24223915 PMCID: PMC3815159 DOI: 10.1371/journal.pone.0079249] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 09/19/2013] [Indexed: 12/26/2022] Open
Abstract
γ-secretase is a large ubiquitously expressed protease complex composed of four core subunits: presenilin, Aph1, PEN-2, and nicastrin. The function of γ-secretase in the cells is to proteolytically cleave various proteins within their transmembrane domains. Presenilin and Aph1 occur as alternative variants belonging to mutually exclusive γ-secretase complexes and providing the complexes with heterogeneous biochemical and physiological properties. γ-secretase is proposed to have a role in the development and progression of cancer and γ-secretase inhibitors are intensively studied for their probable anti-tumor effects in various types of cancer models. Here, we for the first time determined mRNA expression levels of presenilin-1, presenilin-2, Aph1a, Aph1b, PEN-2, and nicastrin in a set of breast cancer tissue samples (N = 55) by quantitative real-time PCR in order to clarify the clinical significance of the expression of different γ-secretase complex components in breast cancer. We found a high positive correlation between the subunit expression levels implying a common regulation of transcription. Our univariate Kaplan-Meier survival analyses established low expression level of γ-secretase complex as a risk factor for breast cancer specific mortality. The tumors expressing low levels of γ-secretase complex were characterized by high histopathological tumor grade, low or no expression of estrogen and progesterone receptors and consequently high probability to fall into the class of triple negative breast cancer tumors. These results may provide novel tools to further categorize breast cancer tumors, especially the highly aggressive and poorly treatable breast cancer type of triple negative cases, and suggest a significant role for γ-secretase in breast cancer.
Collapse
Affiliation(s)
- Hanna M. Peltonen
- Institute of Clinical Medicine, Pathology and Forensic Medicine, University of Eastern Finland, Kuopio, Finland
- Biocenter Kuopio and Cancer Center of Eastern Finland, University of Eastern Finland, Kuopio, Finland
- Imaging Center, Clinical Pathology, Kuopio University Hospital, Kuopio, Finland
- * E-mail:
| | - Annakaisa Haapasalo
- Institute of Clinical Medicine – Neurology, University of Eastern Finland, Kuopio, Finland
| | - Mikko Hiltunen
- Institute of Clinical Medicine – Neurology, University of Eastern Finland, Kuopio, Finland
| | - Vesa Kataja
- Institute of Clinical Medicine, Oncology, University of Eastern Finland, Kuopio, Finland
- Cancer Center, Kuopio University Hospital, Kuopio, Finland
| | - Veli-Matti Kosma
- Institute of Clinical Medicine, Pathology and Forensic Medicine, University of Eastern Finland, Kuopio, Finland
- Biocenter Kuopio and Cancer Center of Eastern Finland, University of Eastern Finland, Kuopio, Finland
- Imaging Center, Clinical Pathology, Kuopio University Hospital, Kuopio, Finland
| | - Arto Mannermaa
- Institute of Clinical Medicine, Pathology and Forensic Medicine, University of Eastern Finland, Kuopio, Finland
- Biocenter Kuopio and Cancer Center of Eastern Finland, University of Eastern Finland, Kuopio, Finland
- Imaging Center, Clinical Pathology, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
23
|
Gai JW, Wahafu W, Hsieh YC, Liu M, Zhang L, Li SW, Zhang B, He Q, Guo H, Jin J. Inhibition of presenilins attenuates proliferation and invasion in bladder cancer cells through multiple pathways. Urol Oncol 2013; 32:36.e19-25. [PMID: 23628311 DOI: 10.1016/j.urolonc.2013.02.014] [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: 11/29/2012] [Revised: 02/21/2013] [Accepted: 02/25/2013] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Presenilin (PS)/γ-secretase is a key protease that initiates various biological processes. We investigated the effect of PS/γ-secretase on the expression and inhibition of urothelial cell carcinoma of bladder (UCB) as a potential alternative therapeutic target for UCB. MATERIALS AND METHODS PS-1 and PS-2 were identified in normal and malignant human bladder transitional cells by immunohistochemistry. We blocked PSs using a PS/γ-secretase inhibitor N-(N-[3,5-difluorophenacetyl]-L-alanyl)-S-phenylglycine-t-butylester (DAPT), and the proliferative and invasive potential of UCB cells SW780, BIU-87, 5637, and T24, and human normal urothelial cell line SV-HUC-1 were analyzed using Western blot, cell viability test, flow cytometry, and transwell assay. All experiments were repeated at least 3 times. RESULTS Human bladder samples of UCB, SW780, BIU-87, 5637, and T24 cells expressed higher PS-1 compared with normal ones. Cell vitality test demonstrated that DAPT attenuated UCB cell proliferation more than SV-HUC-1. Flow cytometry and transwell assay showed that T24 cells were arrested at G1/S checkpoint and its invasive ability was impaired. Western blot assay markedly showed that protein levels of CD44-intracellular domain, insulinlike growth factor-1Rβ, extracellular regulated protein kinase 1/2, cyclin D1, proliferating cell nuclear antigen, and matrix metalloproteinase-9 were downregulated by DAPT, whereas vascular endothelial growth factor receptor-2 and vascular endothelial growth factor-165 were upregulated. CONCLUSIONS Our study revealed that PS-1 might be implicated in the proliferation and invasion of UCB, and that it may serve as a potential therapeutic target for UCB, but further studies are warranted to verify the effects of inhibition of PS/γ-secretase on angiogenesis.
Collapse
Affiliation(s)
- Jun-Wei Gai
- Department of Urology, Peking University First Hospital; Institute of Urology, Peking University, Beijing, China
| | | | - Ya-Ching Hsieh
- Department of Anesthesiology and Intensive Care, Peking University First Hospital, Beijing, China
| | - Miao Liu
- HeDong Center for Disease Control and Prevention, Tianjin, China
| | - Liang Zhang
- Department of Orthopaedic Surgery, Beijing Jishuitan Hospital, Fourth Clinical College of Peking University, Beijing, China
| | - Sheng-Wen Li
- Department of Urology, Tsinghua University First Hospital, Beijing, China
| | - Bei Zhang
- Department of Urology, Peking University First Hospital; Institute of Urology, Peking University, Beijing, China
| | - Qun He
- Department of Urology, Peking University First Hospital; Institute of Urology, Peking University, Beijing, China
| | - Hui Guo
- Department of Urology, Tsinghua University First Hospital, Beijing, China.
| | - Jie Jin
- Department of Urology, Peking University First Hospital; Institute of Urology, Peking University, Beijing, China.
| |
Collapse
|
24
|
Cai J, Qi X, Kociok N, Skosyrski S, Emilio A, Ruan Q, Han S, Liu L, Chen Z, Bowes Rickman C, Golde T, Grant MB, Saftig P, Serneels L, de Strooper B, Joussen AM, Boulton ME. β-Secretase (BACE1) inhibition causes retinal pathology by vascular dysregulation and accumulation of age pigment. EMBO Mol Med 2012; 4:980-991. [PMID: 22903875 PMCID: PMC3491829 DOI: 10.1002/emmm.201101084] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Revised: 06/23/2012] [Accepted: 06/28/2012] [Indexed: 01/18/2023] Open
Abstract
β-Secretase (BACE1) is a major drug target for combating Alzheimer's disease (AD). Here we show that BACE1(-/-) mice develop significant retinal pathology including retinal thinning, apoptosis, reduced retinal vascular density and an increase in the age pigment, lipofuscin. BACE1 expression is highest in the neural retina while BACE2 was greatest in the retinal pigment epithelium (RPE)/choroid. Pigment epithelial-derived factor, a known regulator of γ-secretase, inhibits vascular endothelial growth factor (VEGF)-induced in vitro and in vivo angiogenesis and this is abolished by BACE1 inhibition. Moreover, intravitreal administration of BACE1 inhibitor or BACE1 small interfering RNA (siRNA) increases choroidal neovascularization in mice. BACE1 induces ectodomain shedding of vascular endothelial growth factor receptor 1 (VEGFR1) which is a prerequisite for γ-secretase release of a 100 kDa intracellular domain. The increase in lipofuscin following BACE1 inhibition and RNAI knockdown is associated with lysosomal perturbations. Taken together, our data show that BACE1 plays a critical role in retinal homeostasis and that the use of BACE inhibitors for AD should be viewed with extreme caution as they could lead to retinal pathology and exacerbate conditions such as age-related macular degeneration.
Collapse
Affiliation(s)
- Jun Cai
- Department of Anatomy & Cell Biology, University of FloridaGainesville, FL, USA
| | - Xiaoping Qi
- Department of Anatomy & Cell Biology, University of FloridaGainesville, FL, USA
| | - Norbert Kociok
- Department of Ophthalmology, Charité Universitätsmedizin BerlinBerlin, Germany
| | - Sergej Skosyrski
- Department of Ophthalmology, Charité Universitätsmedizin BerlinBerlin, Germany
| | - Alonso Emilio
- Department of Anatomy & Cell Biology, University of FloridaGainesville, FL, USA
| | - Qing Ruan
- Department of Anatomy & Cell Biology, University of FloridaGainesville, FL, USA
| | - Song Han
- Department of Surgery, University of FloridaGainesville, FL, USA
| | - Li Liu
- Department of Pharmacology & Therapeutics, University of FloridaGainesville, FL, USA
| | - Zhijuan Chen
- Department of Anatomy & Cell Biology, University of FloridaGainesville, FL, USA
| | - Catherine Bowes Rickman
- Department of Ophthalmology & of Cell Biology, Duke University Medical CenterDurham, NC, USA
| | - Todd Golde
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, University of FloridaGainesville, FL, USA
| | - Maria B Grant
- Department of Pharmacology & Therapeutics, University of FloridaGainesville, FL, USA
| | - Paul Saftig
- Biochemical Institute, Christian-Albrecht's UniversityKiel, Germany
| | - Lutgarde Serneels
- Center for Human Genetics and Leuven Institute for Neurodegenerative Diseases (LIND)KU Leuven, Leuven, Belgium
- VIB Center for the Biology of Disease, Vlaams Instituut voor BiotechnologieLeuven, Belgium
| | - Bart de Strooper
- Center for Human Genetics and Leuven Institute for Neurodegenerative Diseases (LIND)KU Leuven, Leuven, Belgium
- VIB Center for the Biology of Disease, Vlaams Instituut voor BiotechnologieLeuven, Belgium
| | - Antonia M Joussen
- Department of Ophthalmology, Charité Universitätsmedizin BerlinBerlin, Germany
| | - Michael E Boulton
- Department of Anatomy & Cell Biology, University of FloridaGainesville, FL, USA
| |
Collapse
|
25
|
Cameron DJ, Galvin C, Alkam T, Sidhu H, Ellison J, Luna S, Ethell DW. Alzheimer's-related peptide amyloid-β plays a conserved role in angiogenesis. PLoS One 2012; 7:e39598. [PMID: 22792182 PMCID: PMC3392248 DOI: 10.1371/journal.pone.0039598] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 05/24/2012] [Indexed: 11/19/2022] Open
Abstract
Alzheimer's disease research has been at an impasse in recent years with lingering questions about the involvement of Amyloid-β (Aβ). Early versions of the amyloid hypothesis considered Aβ something of an undesirable byproduct of APP processing that wreaks havoc on the human neocortex, yet evolutionary conservation--over three hundred million years--indicates this peptide plays an important biological role in survival and reproductive fitness. Here we describe how Aβ regulates blood vessel branching in tissues as varied as human umbilical vein and zebrafish hindbrain. High physiological concentrations of Aβ monomer induced angiogenesis by a conserved mechanism that blocks γ-secretase processing of a Notch intermediate, NEXT, and reduces the expression of downstream Notch target genes. Our findings allude to an integration of signaling pathways that utilize γ-secretase activity, which may have significant implications for our understanding of Alzheimer's pathogenesis vis-à-vis vascular changes that set the stage for ensuing neurodegeneration.
Collapse
Affiliation(s)
- D. Joshua Cameron
- Molecular Neurobiology, Western University of Health Sciences, Pomona, California, United States of America
- College of Optometry, Western University of Health Sciences, Pomona, California, United States of America
| | - Cooper Galvin
- Molecular Neurobiology, Western University of Health Sciences, Pomona, California, United States of America
| | - Tursun Alkam
- Molecular Neurobiology, Western University of Health Sciences, Pomona, California, United States of America
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, California, United States of America
| | - Harpreet Sidhu
- Molecular Neurobiology, Western University of Health Sciences, Pomona, California, United States of America
| | - John Ellison
- Molecular Neurobiology, Western University of Health Sciences, Pomona, California, United States of America
| | - Salvadore Luna
- Molecular Neurobiology, Western University of Health Sciences, Pomona, California, United States of America
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, California, United States of America
| | - Douglas W. Ethell
- Molecular Neurobiology, Western University of Health Sciences, Pomona, California, United States of America
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, California, United States of America
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, United States of America
| |
Collapse
|
26
|
Nuclear translocation and functions of growth factor receptors. Semin Cell Dev Biol 2012; 23:165-71. [DOI: 10.1016/j.semcdb.2011.09.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2011] [Accepted: 09/08/2011] [Indexed: 01/24/2023]
|
27
|
Lee JA, Uhlik MT, Moxham CM, Tomandl D, Sall DJ. Modern phenotypic drug discovery is a viable, neoclassic pharma strategy. J Med Chem 2012; 55:4527-38. [PMID: 22409666 DOI: 10.1021/jm201649s] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Jonathan A Lee
- Department of Quantitative Biology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA.
| | | | | | | | | |
Collapse
|
28
|
Qi X, Cai J, Ruan Q, Liu L, Boye SL, Chen Z, Hauswirth WW, Ryals RC, Shaw L, Caballero S, Grant MB, Boulton ME. γ-Secretase inhibition of murine choroidal neovascularization is associated with reduction of superoxide and proinflammatory cytokines. Invest Ophthalmol Vis Sci 2012; 53:574-85. [PMID: 22205609 DOI: 10.1167/iovs.11-8728] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
PURPOSE This study aimed to determine whether upregulation of γ-secretase could inhibit laser-induced choroidal neovascularization (CNV) and if this was associated with a reduction in both oxidative stress and proinflammatory cytokines. METHODS γ-Secretase, or its catalytic subunit presenilin 1 (PS1), were upregulated by exposure to either pigment epithelial derived factor (PEDF) or an AAV2 vector containing a PS1 gene driven by a vascular endothelial-cadherin promoter. Retinal endothelial cells were infected with AAV2 or exposed to PEDF in the presence or absence of VEGF and in vitro angiogenesis determined. Mouse eyes either received intravitreal injection of PEDF, DAPT (a γ-secretase inhibitor) or PEDF + DAPT at the time of laser injury, or AAV2 infection 3 weeks before receiving laser burns. Lesion volume was determined 14 days post laser injury. Superoxide generation, antioxidant activity and the production of proinflammatory mediators were assessed. Knockdown of γ-secretase was achieved using siRNA. RESULTS γ-Secretase upregulation and PS1 overexpression suppressed VEGF-induced in vitro angiogenesis and in vivo laser-induced CNV. This was associated with a reduction in the expression of VEGF and angiogenin 1 together with reduced superoxide anion generation and an increase in MnSOD compared with untreated CNV eyes. PS1 overexpression reduced proinflammatory factors and microglial activation in eyes with CNV compared with control. siRNA inhibition of γ-secretase resulted in increased angiogenesis. CONCLUSIONS γ-Secretase, and in particular PS1 alone, are potent regulators of angiogenesis and this is due in part to stabilizing endogenous superoxide generation and reducing proinflammatory cytokine expression during CNV.
Collapse
Affiliation(s)
- Xiaoping Qi
- Departments of Anatomy and Cell Biology, University of Florida, Gainesville, Florida 32610-0235, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Cai J, Chen Z, Ruan Q, Han S, Liu L, Qi X, Boye SL, Hauswirth WW, Grant MB, Boulton ME. γ-Secretase and presenilin mediate cleavage and phosphorylation of vascular endothelial growth factor receptor-1. J Biol Chem 2011; 286:42514-42523. [PMID: 22016384 PMCID: PMC3234916 DOI: 10.1074/jbc.m111.296590] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Revised: 10/14/2011] [Indexed: 01/18/2023] Open
Abstract
We have reported previously that pigment epithelium-derived factor (PEDF) can, via γ-secretase-mediated events, inhibit VEGF-induced angiogenesis in microvascular endothelial cells by both (a) cleavage and intracellular translocation of a C-terminal fragment of VEGF receptor-1 (VEGFR1) and (b) inhibition of VEGF-induced phosphorylation of VEGFR1. Using site-direct mutagenesis and transfection of wild type and mutated receptors into endothelial cells, we showed that transmembrane cleavage of VEGFR1 occurs at valine 767 and that a switch from valine to alanine at this position prevented cleavage and formation of a VEGFR1 intracellular fragment. Using siRNA to selectively knock down protein-tyrosine phosphatases (PTPs) in endothelial cells, we demonstrated that vascular endothelial PTP is responsible for dephosphorylation of activated VEGFR1. PEDF up-regulation of full-length presenilin 1 (Fl.PS1) facilitated the association of vascular endothelial PTP and VEGFR1. Knockdown of Fl.PS1 prevented dephosphorylation of VEGFR1, whereas up-regulation of Fl.PS1 stimulated VEGFR1 dephosphorylation. Fl.PS1 associated with VEGFR1 within 15 min after PEDF treatment. In conclusion, we determined the PEDF-mediated events responsible for VEGFR1 signaling and identified full-length presenilin as a critical adaptor molecule in the dephosphorylation of VEGFR1. This greater understanding of the regulation of VEGFR1 signaling will help identify novel anti-VEGF therapeutic strategies.
Collapse
Affiliation(s)
- Jun Cai
- Departments of Anatomy and Cell Biology, University of Florida, Gainesville, Florida 32610-0235
| | - Zhijuan Chen
- Departments of Anatomy and Cell Biology, University of Florida, Gainesville, Florida 32610-0235
| | - Qing Ruan
- Departments of Anatomy and Cell Biology, University of Florida, Gainesville, Florida 32610-0235
| | - Song Han
- Department of Surgery, University of Florida, Gainesville, Florida 32610-0235
| | - Li Liu
- Departments of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida 32610-0235
| | - Xiaoping Qi
- Departments of Anatomy and Cell Biology, University of Florida, Gainesville, Florida 32610-0235
| | - Sanford L Boye
- Department of Ophthalmology, University of Florida, Gainesville, Florida 32610-0235
| | - William W Hauswirth
- Department of Ophthalmology, University of Florida, Gainesville, Florida 32610-0235
| | - Maria B Grant
- Departments of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida 32610-0235
| | - Michael E Boulton
- Departments of Anatomy and Cell Biology, University of Florida, Gainesville, Florida 32610-0235.
| |
Collapse
|
30
|
Le Moan N, Houslay DM, Christian F, Houslay MD, Akassoglou K. Oxygen-dependent cleavage of the p75 neurotrophin receptor triggers stabilization of HIF-1α. Mol Cell 2011; 44:476-90. [PMID: 22055192 PMCID: PMC3212815 DOI: 10.1016/j.molcel.2011.08.033] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Revised: 05/23/2011] [Accepted: 08/15/2011] [Indexed: 12/23/2022]
Abstract
Homeostatic control of oxygen availability allows cells to survive oxygen deprivation. Although the transcription factor hypoxia-inducible factor 1α (HIF-1α) is the main regulator of the hypoxic response, the upstream mechanisms required for its stabilization remain elusive. Here, we show that p75 neurotrophin receptor (p75(NTR)) undergoes hypoxia-induced γ-secretase-dependent cleavage to provide a positive feed-forward mechanism required for oxygen-dependent HIF-1α stabilization. The intracellular domain of p75(NTR) directly interacts with the evolutionarily conserved zinc finger domains of the E3 RING ubiquitin ligase Siah2 (seven in absentia homolog 2), which regulates HIF-1α degradation. p75(NTR) stabilizes Siah2 by decreasing its auto-ubiquitination. Genetic loss of p75(NTR) dramatically decreases Siah2 abundance, HIF-1α stabilization, and induction of HIF-1α target genes in hypoxia. p75(NTR-/-) mice show reduced HIF-1α stabilization, vascular endothelial growth factor (VEGF) expression, and neoangiogenesis after retinal hypoxia. Thus, hypoxia-induced intramembrane proteolysis of p75(NTR) constitutes an apical oxygen-dependent mechanism to control the magnitude of the hypoxic response.
Collapse
Affiliation(s)
- Natacha Le Moan
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | - Daniel M. Houslay
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | - Frank Christian
- Molecular Pharmacology Group, Biochemistry & Molecular Biology, University of Glasgow, Glasgow G12 8QQ, Scotland, UK
| | - Miles D. Houslay
- Molecular Pharmacology Group, Biochemistry & Molecular Biology, University of Glasgow, Glasgow G12 8QQ, Scotland, UK
| | - Katerina Akassoglou
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
- Department of Neurology, University of California, San Francisco, CA 94158, USA
| |
Collapse
|
31
|
Merkwitz C, Lochhead P, Tsikolia N, Koch D, Sygnecka K, Sakurai M, Spanel-Borowski K, Ricken AM. Expression of KIT in the ovary, and the role of somatic precursor cells. ACTA ACUST UNITED AC 2011; 46:131-84. [DOI: 10.1016/j.proghi.2011.09.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
32
|
Arima S, Nishiyama K, Ko T, Arima Y, Hakozaki Y, Sugihara K, Koseki H, Uchijima Y, Kurihara Y, Kurihara H. Angiogenic morphogenesis driven by dynamic and heterogeneous collective endothelial cell movement. Development 2011; 138:4763-76. [DOI: 10.1242/dev.068023] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Angiogenesis is a complex process, which is accomplished by reiteration of modules such as sprouting, elongation and bifurcation, that configures branching vascular networks. However, details of the individual and collective behaviors of vascular endothelial cells (ECs) during angiogenic morphogenesis remain largely unknown. Herein, we established a time-lapse imaging and computer-assisted analysis system that quantitatively characterizes behaviors in sprouting angiogenesis. Surprisingly, ECs moved backwards and forwards, overtaking each other even at the tip, showing an unknown mode of collective cell movement with dynamic ‘cell-mixing’. Mosaic analysis, which enabled us to monitor the behavior of individual cells in a multicellular structure, confirmed the ‘cell-mixing’ phenomenon of ECs that occurs at the whole-cell level. Furthermore, an in vivo EC-tracking analysis revealed evidence of cell-mixing and overtaking at the tip in developing murine retinal vessels. In parametrical analysis, VEGF enhanced tip cell behavior and directed EC migration at the stalk during branch elongation. These movements were counter-regulated by EC-EC interplay via γ-secretase-dependent Dll4-Notch signaling, and might be promoted by EC-mural cell interplay. Finally, multiple regression analysis showed that these molecule-mediated tip cell behaviors and directed EC migration contributed to effective branch elongation. Taken together, our findings provide new insights into the individual and collective EC movements driving angiogenic morphogenesis. The methodology used for this analysis might serve to bridge the gap in our understanding between individual cell behavior and branching morphogenesis.
Collapse
Affiliation(s)
- Satoshi Arima
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Koichi Nishiyama
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Toshiyuki Ko
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Yuichiro Arima
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Yuji Hakozaki
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Kei Sugihara
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Hiroaki Koseki
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Yasunobu Uchijima
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Yukiko Kurihara
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Hiroki Kurihara
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| |
Collapse
|
33
|
Cai J, Wu L, Qi X, Li Calzi S, Caballero S, Shaw L, Ruan Q, Grant MB, Boulton ME. PEDF regulates vascular permeability by a γ-secretase-mediated pathway. PLoS One 2011; 6:e21164. [PMID: 21695048 PMCID: PMC3117873 DOI: 10.1371/journal.pone.0021164] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Accepted: 05/21/2011] [Indexed: 01/22/2023] Open
Abstract
Increased vascular permeability is an inciting event in many vascular complications including diabetic retinopathy. We have previously reported that pigment epithelium-derived factor (PEDF) is able to inhibit vascular endothelial growth factor (VEGF)-induced angiogenesis through a novel γ-secretase-dependent pathway. In this study, we asked whether inhibition of VEGF-induced permeability by PEDF is also γ-secretase-mediated and to dissect the potential mechanisms involved. Vascular permeability was assessed in vitro by measuring transendothelial resistance and paracellular permeability to dextran and in vivo by following leakage of intravenous FITC-labelled albumin into the retina in the presence or absence of VEGF and PEDF in varying combinations. Experiments were undertaken in the presence or absence of a γ-secretase inhibitor. To assess junctional integrity immunohistochemistry for the adherens junction (AJ) proteins, VE-cadherin and β-catenin, and the tight junction (TJ) protein, claudin-5 was undertaken using cultured cells and flat mount retinas. Protein expression and the association between AJ proteins, VEGF receptors (VEGFRs) and γ-secretase constituents were determined by immunoprecipitation and Western Blot analysis. In selected experiments the effect of hypoxia on junctional integrity was also assessed. PEDF inhibition of VEGF-induced permeability, both in cultured microvascular endothelial cell monolayers and in vivo in the mouse retinal vasculature, is mediated by γ-secretase. PEDF acted by a) preventing dissociation of AJ and TJ proteins and b) regulating both the association of VEGF receptors with AJ proteins and the subsequent phosphorylation of the AJ proteins, VE-cadherin and β-catenin. Association of γ-secretase with AJ proteins appears to be critical in the regulation of vascular permeability. Although hypoxia increased VEGFR expression there was a significant dissociation of VEGFR from AJ proteins. In conclusion, PEDF regulates VEGF-induced vascular permeability via a novel γ-secretase dependent pathway and targeting downstream effectors of PEDF action may represent a promising therapeutic strategy for preventing or ameliorating increased vascular permeability.
Collapse
Affiliation(s)
- Jun Cai
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, Florida, United States of America
| | - Lin Wu
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, Florida, United States of America
| | - Xiaoping Qi
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, Florida, United States of America
| | - Sergio Li Calzi
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida, United States of America
| | - Sergio Caballero
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida, United States of America
| | - Lynn Shaw
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida, United States of America
| | - Qing Ruan
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, Florida, United States of America
| | - Maria B. Grant
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida, United States of America
| | - Michael E. Boulton
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, Florida, United States of America
- * E-mail:
| |
Collapse
|
34
|
Whyte JL, Ball SG, Shuttleworth CA, Brennan K, Kielty CM. Density of human bone marrow stromal cells regulates commitment to vascular lineages. Stem Cell Res 2011; 6:238-50. [PMID: 21420373 PMCID: PMC3223522 DOI: 10.1016/j.scr.2011.02.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2010] [Revised: 02/04/2011] [Accepted: 02/09/2011] [Indexed: 02/06/2023] Open
Abstract
Mechanisms underlying the vascular differentiation of human bone marrow stromal cells (HBMSCs) and their contribution to neovascularisation are poorly understood. We report the essential role of cell density-induced signals in directing HBMSCs along endothelial or smooth muscle lineages. Plating HBMSCs at high density rapidly induced Notch signaling, which initiated HBMSC commitment to a vascular progenitor cell population expressing markers for both vascular lineages. Notch also induced VEGF-A, which inhibited vascular smooth muscle commitment while consolidating differentiation to endothelial cells with cobblestone morphology and characteristic endothelial markers and functions. These mechanisms can be exploited therapeutically to regulate HBMSCs during neovascularisation.
Collapse
Affiliation(s)
| | | | | | | | - Cay M. Kielty
- Corresponding author at: Faculty of Life Sciences, University of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK. Fax: +44 161 275 5082.
| |
Collapse
|
35
|
Gordon-Keylock SAM, Jackson M, Huang C, Samuel K, Axton RA, Oostendorp RAJ, Taylor H, Wilson J, Forrester LM. Induction of hematopoietic differentiation of mouse embryonic stem cells by an AGM-derived stromal cell line is not further enhanced by overexpression of HOXB4. Stem Cells Dev 2010; 19:1687-98. [PMID: 20184433 DOI: 10.1089/scd.2009.0467] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hematopoietic differentiation of embryonic stem (ES) cells can be enhanced by co-culture with stromal cells derived from hematopoietic tissues and by overexpression of the transcription factor HOXB4. In this study, we compare the hematopoietic inductive effects of stromal cell lines derived from different subregions of the embryonic aorta-gonad-mesonephros tissue with the commonly used OP9 stromal cell line and with HOXB4 activation. We show that stromal cell lines derived from the aorta and surrounding mesenchyme (AM) act at an earlier stage of the differentiation process compared with the commonly used OP9 stromal cells. AM stromal cells were able to promote the further differentiation of isolated brachyury-GFP(+) mesodermal cells into hematopoietic progenitors, whereas the OP9 stromal cells could not support the differentiation of these cells. Co-culture and analyses of individual embryoid bodies support the hypothesis that the AM stromal cell lines could enhance the de novo production of hematopoietic progenitors, lending support to the idea that AM stromal cells might act on prehematopoietic mesoderm. The induction level observed for AM stromal cells was comparable to HOXB4 activation, but no additive effect was observed when these 2 inductive strategies were combined. Addition of a γ-secretase inhibitor reduced the inductive effects of both the stromal cell line and HOXB4, providing clues to possible shared molecular mechanisms.
Collapse
Affiliation(s)
- Sabrina A M Gordon-Keylock
- MRC Centre for Regenerative Medicine, John Hughes Bennett Laboratory, Queens Medical Research Institute, Edinburgh, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Bae YH, Park HJ, Kim SR, Kim JY, Kang Y, Kim JA, Wee HJ, Kageyama R, Jung JS, Bae MK, Bae SK. Notch1 mediates visfatin-induced FGF-2 up-regulation and endothelial angiogenesis. Cardiovasc Res 2010; 89:436-45. [PMID: 20817637 DOI: 10.1093/cvr/cvq276] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
AIMS Our aims were to determine the role of Notch1 in mediating visfatin-induced angiogenesis and to explore potential target genes involved. METHODS AND RESULTS Inhibition of Notch signalling attenuated visfatin-induced angiogenesis in vitro, ex vivo, and in vivo. Visfatin increased γ-secretase activity, Notch1 cleavage and activation, and Hes1 gene induction. Visfatin also stimulated fibroblast growth factor-2 (FGF-2) gene expression in a Notch1-dependent manner. Enforced expression of active Notch1 intracellular domain increased FGF-2 protein levels and stimulated endothelial tube formation, whereas blocking Notch1 signalling or knockdown of Notch1 by small interfering RNA suppressed visfatin-induced FGF-2 up-regulation and angiogenesis. Reporter analysis of FGF-2 promoter revealed the presence of CSL (CBF-1, suppressor of hairless, LAG-1)-binding site, and chromatin immunoprecipitation analysis demonstrated the binding of Notch1-CSL complex to this site in response to visfatin. CONCLUSION Our data provide the first example of Notch1-dependent endothelial FGF-2 induction by visfatin and of Notch1 activation in visfatin-stimulated endothelial angiogenesis, suggesting that the signalling axis of visfatin/Notch1/angiogenic factors like FGF-2 might be a valuable target for pathological angiogenesis.
Collapse
Affiliation(s)
- Yun-Hee Bae
- Department of Physiology, School of Medicine, Yangsan Campus of Pusan National University, Yangsan 626-870, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Moustakas A, Kreisl TN. New treatment options in the management of glioblastoma multiforme: a focus on bevacizumab. Onco Targets Ther 2010; 3:27-38. [PMID: 20616955 PMCID: PMC2895775 DOI: 10.2147/ott.s5307] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2010] [Indexed: 12/21/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common malignant primary brain tumor in adults and carries the poorest prognosis. Despite recent progress in molecular biology, neuro-imaging and neuro-surgical care, the management of patients with GBM continues to harbor significant challenges. Survival after diagnosis is poor even with the most aggressive approach using multimodality therapy. Although the etiology of malignant gliomas is not known, the dependency of tumor growth on angiogenesis has identified this pathway as a promising therapeutic target. Bevacizumab was the first antiangiogenic therapy approved for use in cancer and received accelerated Food and Drug Administration approval for the treatment of recurrent GBM in 2009, the first new drug for this disease in over a decade. This review describes the rationale behind the treatment of GBM with bevacizumab. The pharmacology, efficacy, safety and tolerability of bevacizumab will also be reviewed.
Collapse
Affiliation(s)
- Argirios Moustakas
- National Cancer Institute, Neuro-Oncology Branch, National Institutes of Health, Bethesda, Maryland, USA
| | | |
Collapse
|
38
|
Wu F, Schweizer C, Rudinskiy N, Taylor DM, Kazantsev A, Luthi-Carter R, Fraering PC. Novel gamma-secretase inhibitors uncover a common nucleotide-binding site in JAK3, SIRT2, and PS1. FASEB J 2010; 24:2464-74. [PMID: 20237298 DOI: 10.1096/fj.09-148031] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Gamma-secretase is an intramembrane-cleaving protease responsible for the final proteolytic event in the production of the amyloid-beta peptides (Abeta) implicated in Alzheimer's disease (AD). Inhibition of gamma-secretase activity is thus an attractive therapeutic strategy to slow down the pathogenesis of AD. Drugs often target more than one biomolecule because of conserved 3-dimensional structures in prospective protein binding sites. We have capitalized on this phenomenon of nature to identify new gamma-secretase inhibitors. Here we show that 2-hydroxy naphthyl derivatives, a previously identified subclass of NAD(+) analog inhibitors of sirtuin 2 (SIRT2), are direct gamma-secretase inhibitors. Subsequent structure-activity relationship studies further showed that 2-hydroxy-1-naphthaldehyde is the minimal pharmacophore for gamma-secretase inhibition. In evaluating target protein determinants of inhibition, we identified a common GXG signature nucleotide-binding site (NBS) shared by the gamma-secretase subunit presenilin-1 C-terminal fragment (PS1-CTF), SIRT2, and Janus kinase 3 (JAK3). Because a detailed 3-dimensional structure of gamma-secretase is beyond our knowledge, we took advantage of the known crystal structure of human JAK3 to model the NBS of the PS1-CTF, which includes the catalytic residue D385. Our results suggest that the flexible PS1-CTF (381)LGLG(384) loop comprises a substrate-docking site capable of recognizing specifically different gamma-secretase substrates.
Collapse
Affiliation(s)
- Fang Wu
- Laboratory of Molecular and Cellular Biology of Alzheimer's Disease, Brain Mind Institute and School of Life Sciences, Swiss Federal Institute of Technology, Lausanne, Switzerland
| | | | | | | | | | | | | |
Collapse
|
39
|
Kim WY, Lee HY. Brain angiogenesis in developmental and pathological processes: mechanism and therapeutic intervention in brain tumors. FEBS J 2009; 276:4653-64. [PMID: 19664069 PMCID: PMC2847309 DOI: 10.1111/j.1742-4658.2009.07177.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Formation of new blood vessels is required for the growth and metastasis of all solid tumors. New blood vessels are established in tumors mainly through angiogenesis. Brain tumors in particular are highly angiogenic. Therefore, interventions designed to prevent angiogenesis may be effective at controlling brain tumors. Indeed, many recent findings from preclinical and clinical studies of antiangiogenic therapy for brain tumors have shown that it is a promising approach to managing this deadly disease, especially when combined with other cytotoxic treatments. In this minireview, we summarize the basic characteristics of brain tumor angiogenesis and the role of known angiogenic factors in regulating this angiogenesis, which may be targets of antiangiogenic therapy. We also discuss the current status of antiangiogenic therapy for brain tumors, the suggested mechanisms of this therapy and the limitations of this strategy.
Collapse
Affiliation(s)
- Woo-Young Kim
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | | |
Collapse
|
40
|
Branching morphogenesis and antiangiogenesis candidates: tip cells lead the way. Nat Rev Clin Oncol 2009; 6:315-26. [DOI: 10.1038/nrclinonc.2009.64] [Citation(s) in RCA: 156] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
41
|
De Smet F, Segura I, De Bock K, Hohensinner PJ, Carmeliet P. Mechanisms of Vessel Branching. Arterioscler Thromb Vasc Biol 2009; 29:639-49. [DOI: 10.1161/atvbaha.109.185165] [Citation(s) in RCA: 283] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Filopodia, “the fingers that do the walking,” have been identified on endothelial cells at the tip of sprouting vessels for half a century, but the key role of the tip cell in vessel branching has been recognized only in the past few years. A model is emerging, whereby tip cells lead the way in a branching vessel, stalk cells elongate the sprout, and a very recently discovered phalanx cell ensures quiescence and perfusion of the newly formed branch. Recent genetic studies have shed light on the molecular signature of these distinct endothelial phenotypes; this provides a novel conceptual framework of how vessel morphogenesis occurs. Here, we will discuss the molecular candidates that participate in the decision of endothelial cells to adapt these distinct fates and highlight the emerging insights on how these cells send out filopodia while navigating.
Collapse
Affiliation(s)
| | | | - Katrien De Bock
- From the Vesalius Research Center, VIB, K.U. Leuven, Belgium
| | | | - Peter Carmeliet
- From the Vesalius Research Center, VIB, K.U. Leuven, Belgium
| |
Collapse
|