125 results on '"Guo-Hua Fong"'
Search Results
2. Inhibition of FLT1 ameliorates muscular dystrophy phenotype by increased vasculature in a mouse model of Duchenne muscular dystrophy.
- Author
-
Mayank Verma, Yuko Shimizu-Motohashi, Yoko Asakura, James P Ennen, Jennifer Bosco, Zhiwei Zhou, Guo-Hua Fong, Serene Josiah, Dennis Keefe, and Atsushi Asakura
- Subjects
Genetics ,QH426-470 - Abstract
Duchenne muscular dystrophy (DMD) is an X-linked recessive genetic disease in which the dystrophin coding for a membrane stabilizing protein is mutated. Recently, the vasculature has also shown to be perturbed in DMD and DMD model mdx mice. Recent DMD transcriptomics revealed the defects were correlated to a vascular endothelial growth factor (VEGF) signaling pathway. To reveal the relationship between DMD and VEGF signaling, mdx mice were crossed with constitutive (CAGCreERTM:Flt1LoxP/LoxP) and endothelial cell-specific conditional gene knockout mice (Cdh5CreERT2:Flt1LoxP/LoxP) for Flt1 (VEGFR1) which is a decoy receptor for VEGF. Here, we showed that while constitutive deletion of Flt1 is detrimental to the skeletal muscle function, endothelial cell-specific Flt1 deletion resulted in increased vascular density, increased satellite cell number and improvement in the DMD-associated phenotype in the mdx mice. These decreases in pathology, including improved muscle histology and function, were recapitulated in mdx mice given anti-FLT1 peptides or monoclonal antibodies, which blocked VEGF-FLT1 binding. The histological and functional improvement of dystrophic muscle by FLT1 blockade provides a novel pharmacological strategy for the potential treatment of DMD.
- Published
- 2019
- Full Text
- View/download PDF
3. Hypoxia inducible factor-2α regulates the development of retinal astrocytic network by maintaining adequate supply of astrocyte progenitors.
- Author
-
Li-Juan Duan, Kotaro Takeda, and Guo-Hua Fong
- Subjects
Medicine ,Science - Abstract
Here we investigate the role of hypoxia inducible factor (HIF)-2α in coordinating the development of retinal astrocytic and vascular networks. Three Cre mouse lines were used to disrupt floxed Hif-2α, including Rosa26(CreERT2), Tie2(Cre), and GFAP(Cre). Global Hif-2α disruption by Rosa26(CreERT2) led to reduced astrocytic and vascular development in neonatal retinas, whereas endothelial disruption by Tie2(Cre) had no apparent effects. Hif-2α deletion in astrocyte progenitors by GFAP(Cre) significantly interfered with the development of astrocytic networks, which failed to reach the retinal periphery and were incapable of supporting vascular development. Perplexingly, the abundance of strongly GFAP(+) mature astrocytes transiently increased at P0 before they began to lag behind the normal controls by P3. Pax2(+) and PDGFRα(+) astrocytic progenitors and immature astrocytes were dramatically diminished at all stages examined. Despite decreased number of astrocyte progenitors, their proliferation index or apoptosis was not altered. The above data can be reconciled by proposing that HIF-2α is required for maintaining the supply of astrocyte progenitors by slowing down their differentiation into non-proliferative mature astrocytes. HIF-2α deficiency in astrocyte progenitors may accelerate their differentiation into astrocytes, a change which greatly interferes with the replenishment of astrocyte progenitors due to insufficient time for proliferation. Rapidly declining progenitor supply may lead to premature cessation of astrocyte development. Given that HIF-2α protein undergoes oxygen dependent degradation, an interesting possibility is that retinal blood vessels may regulate astrocyte differentiation through their oxygen delivery function. While our findings support the consensus that retinal astrocytic template guides vascular development, they also raise the possibility that astrocytic and vascular networks may mutually regulate each other's development, mediated at least in part by HIF-2α.
- Published
- 2014
- Full Text
- View/download PDF
4. Correction: Prostate Specific Membrane Antigen (PSMA) Regulates Angiogenesis Independently of VEGF during Ocular Neovascularization.
- Author
-
Christina L. Grant, Leslie A. Caromile, Khayyam Durrani, M. Mamunur Rahman, Kevin P. Claffey, Guo-Hua Fong, and Linda H. Shapiro
- Subjects
Medicine ,Science - Published
- 2012
- Full Text
- View/download PDF
5. Prostate specific membrane antigen (PSMA) regulates angiogenesis independently of VEGF during ocular neovascularization.
- Author
-
Christina L Grant, Leslie A Caromile, Vivienne Ho, Khayyam Durrani, M Mamunur Rahman, Kevin P Claffey, Guo-Hua Fong, and Linda H Shapiro
- Subjects
Medicine ,Science - Abstract
Aberrant growth of blood vessels in the eye forms the basis of many incapacitating diseases and currently the majority of patients respond to anti-angiogenic therapies based on blocking the principal angiogenic growth factor, vascular endothelial growth factor (VEGF). While highly successful, new therapeutic targets are critical for the increasing number of individuals susceptible to retina-related pathologies in our increasingly aging population. Prostate specific membrane antigen (PSMA) is a cell surface peptidase that is absent on normal tissue vasculature but is highly expressed on the neovasculature of most solid tumors, where we have previously shown to regulate angiogenic endothelial cell invasion. Because pathologic angiogenic responses are often triggered by distinct signals, we sought to determine if PSMA also contributes to the pathologic angiogenesis provoked by hypoxia of the retina, which underlies many debilitating retinopathies.Using a mouse model of oxygen-induced retinopathy, we found that while developmental angiogenesis is normal in PSMA null mice, hypoxic challenge resulted in decreased retinal vascular pathology when compared to wild type mice as assessed by avascular area and numbers of vascular tufts/glomeruli. The vessels formed in the PSMA null mice were more organized and highly perfused, suggesting a more 'normal' phenotype. Importantly, the decrease in angiogenesis was not due to an impaired hypoxic response as levels of pro-angiogenic factors are comparable; indicating that PSMA regulation of angiogenesis is independent of VEGF. Furthermore, both systemic and intravitreal administration of a PSMA inhibitor in wild type mice undergoing OIR mimicked the PSMA null phenotype resulting in improved retinal vasculature.Our data indicate that PSMA plays a VEGF-independent role in retinal angiogenesis and that the lack of or inhibition of PSMA may represent a novel therapeutic strategy for treatment of angiogenesis-based ocular diseases.
- Published
- 2012
- Full Text
- View/download PDF
6. Endothelial HIF2α suppresses retinal angiogenesis in neonatal mice by upregulating NOTCH signaling.
- Author
-
Li-Juan Duan, Yida Jiang, and Guo-Hua Fong
- Subjects
NEOVASCULARIZATION ,NOTCH genes ,OXYGEN detectors ,GENE targeting ,CHEMICAL inhibitors ,RETROLENTAL fibroplasia ,ENDOTHELIAL cells - Abstract
Prolyl hydroxylase domain (PHD) proteins are oxygen sensors that use intracellular oxygen as a substrate to hydroxylate hypoxiainducible factor (HIF) α proteins, routing them for polyubiquitylation and proteasomal degradation. Typically, HIFα accumulation in hypoxic or PHD-deficient tissues leads to upregulated angiogenesis. Here, we report unexpected retinal phenotypes associated with endothelial cell (EC)-specific gene targeting of Phd2 (Egln1) and Hif2alpha (Epas1). EC-specific Phd2 disruption suppressed retinal angiogenesis, despite HIFα accumulation and VEGFA upregulation. Suppressed retinal angiogenesis was observed both in development and in the oxygeninduced retinopathy (OIR) model. On the other hand, EC-specific deletion of Hif1alpha (Hif1a), Hif2alpha, or both did not affect retinal vascular morphogenesis. Strikingly, retinal angiogenesis appeared normal in mice double-deficient for endothelial PHD2 and HIF2α. In PHD2-deficient retinal vasculature, delta-like 4 (DLL4, a NOTCH ligand) and HEY2 (a NOTCH target) were upregulated by HIF2αdependent mechanisms. Inhibition of NOTCH signaling by a chemical inhibitor or DLL4 antibody partially rescued retinal angiogenesis. Taken together, our data demonstrate that HIF2α accumulation in retinal ECs inhibits rather than stimulates retinal angiogenesis, in part by upregulating DLL4 expression and NOTCH signaling. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
7. Data from PanIN-Specific Regulation of Wnt Signaling by HIF2α during Early Pancreatic Tumorigenesis
- Author
-
Farzad Esni, Guo-Hua Fong, George K. Gittes, Michael T. Lotze, Satdarshan P.S. Monga, Douglas J. Hartman, Emily Wickline, Volker Fendrich, Julie A. Rhodes, Li-Juan Duan, and Angela Criscimanna
- Abstract
Hypoxia promotes angiogenesis, proliferation, invasion, and metastasis of pancreatic cancer. Essentially, all studies of the hypoxia pathway in pancreatic cancer research to date have focused on fully malignant tumors or cancer cell lines, but the potential role of hypoxia inducible factors (HIF) in the progression of premalignant lesions has not been critically examined. Here, we show that HIF2α is expressed early in pancreatic lesions both in human and in a mouse model of pancreatic cancer. HIF2α is a potent oncogenic stimulus, but its role in Kras-induced pancreatic neoplasia has not been discerned. We used the Ptf1aCre transgene to activate KrasG12D and delete Hif2α solely within the pancreas. Surprisingly, loss of Hif2α in this model led to markedly higher, rather than reduced, number of low-grade pancreatic intraepithelial neoplasia (mPanIN) lesions. These lesions, however, failed to progress to high-grade mPanINs, and displayed exclusive loss of β-catenin and SMAD4. The relationship among HIF2α, β-catenin, and Smad4 was further confirmed in vitro, where silencing of Hif2α resulted in reduced β-catenin and Smad4 transcript levels. Thus, with oncogenic Ras expressed in the pancreas, HIF2α modulates Wnt-signaling during mPanIN progression by maintaining appropriate levels of both Smad4 and β-catenin. Cancer Res; 73(15); 4781–90. ©2013 AACR.
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementry Figure from PanIN-Specific Regulation of Wnt Signaling by HIF2α during Early Pancreatic Tumorigenesis
- Author
-
Farzad Esni, Guo-Hua Fong, George K. Gittes, Michael T. Lotze, Satdarshan P.S. Monga, Douglas J. Hartman, Emily Wickline, Volker Fendrich, Julie A. Rhodes, Li-Juan Duan, and Angela Criscimanna
- Abstract
Supplementry Figure 4 - PDF file 812K, HIF2� promotes B-catenin and Smad4 expression
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Figure 2 from PanIN-Specific Regulation of Wnt Signaling by HIF2α during Early Pancreatic Tumorigenesis
- Author
-
Farzad Esni, Guo-Hua Fong, George K. Gittes, Michael T. Lotze, Satdarshan P.S. Monga, Douglas J. Hartman, Emily Wickline, Volker Fendrich, Julie A. Rhodes, Li-Juan Duan, and Angela Criscimanna
- Abstract
Supplementary Figure 2 - PDF file 2100K, Normal pancreas development in Ptf1Cre;Hif2�f/f mice
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Figure 5 from PanIN-Specific Regulation of Wnt Signaling by HIF2α during Early Pancreatic Tumorigenesis
- Author
-
Farzad Esni, Guo-Hua Fong, George K. Gittes, Michael T. Lotze, Satdarshan P.S. Monga, Douglas J. Hartman, Emily Wickline, Volker Fendrich, Julie A. Rhodes, Li-Juan Duan, and Angela Criscimanna
- Abstract
Supplementary Figure 5 - PDF file 5613 K, Loss of Smad4 in PKH2 mPanINs
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Materials and Methods from PanIN-Specific Regulation of Wnt Signaling by HIF2α during Early Pancreatic Tumorigenesis
- Author
-
Farzad Esni, Guo-Hua Fong, George K. Gittes, Michael T. Lotze, Satdarshan P.S. Monga, Douglas J. Hartman, Emily Wickline, Volker Fendrich, Julie A. Rhodes, Li-Juan Duan, and Angela Criscimanna
- Abstract
Supplementary Materials and Methods- PDF file 100K, Additional experimental procedures including Generation of Hif2�-floxed mouse line, Tissue processing and Immunostaining, Western Blot, and Small Interfering RNA transfection
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Figure 1 from PanIN-Specific Regulation of Wnt Signaling by HIF2α during Early Pancreatic Tumorigenesis
- Author
-
Farzad Esni, Guo-Hua Fong, George K. Gittes, Michael T. Lotze, Satdarshan P.S. Monga, Douglas J. Hartman, Emily Wickline, Volker Fendrich, Julie A. Rhodes, Li-Juan Duan, and Angela Criscimanna
- Abstract
Supplementary Figure 1 - PDF file 2170K, Generation of the Hif2�-floxed strain, and validation of Hif2� inactivation in the pancreas
- Published
- 2023
- Full Text
- View/download PDF
13. Supplementary Tables S1-S2 from PanIN-Specific Regulation of Wnt Signaling by HIF2α during Early Pancreatic Tumorigenesis
- Author
-
Farzad Esni, Guo-Hua Fong, George K. Gittes, Michael T. Lotze, Satdarshan P.S. Monga, Douglas J. Hartman, Emily Wickline, Volker Fendrich, Julie A. Rhodes, Li-Juan Duan, and Angela Criscimanna
- Abstract
Supplementary Tables S1-S2 - PDF file 73K, List of primary antibodies, along with the sources and working dilutions (S1); primers used in RT-PCR analyses (S2)
- Published
- 2023
- Full Text
- View/download PDF
14. Supplementary Figure and Table Legends from PanIN-Specific Regulation of Wnt Signaling by HIF2α during Early Pancreatic Tumorigenesis
- Author
-
Farzad Esni, Guo-Hua Fong, George K. Gittes, Michael T. Lotze, Satdarshan P.S. Monga, Douglas J. Hartman, Emily Wickline, Volker Fendrich, Julie A. Rhodes, Li-Juan Duan, and Angela Criscimanna
- Abstract
Supplementary Figure and Table Legends�- PDF file 93K, Legend for Supplemental Tables S1-S2 and Supplemental Figures S1-S5
- Published
- 2023
- Full Text
- View/download PDF
15. Oxygen-sensing mechanisms in development and tissue repair
- Author
-
Yida Jiang, Li-Juan Duan, and Guo-Hua Fong
- Subjects
Oxygen ,Mice ,Ubiquitination ,Animals ,Embryonic Development ,Embryo, Mammalian ,Hypoxia-Inducible Factor 1, alpha Subunit ,Molecular Biology ,Primer ,Developmental Biology - Abstract
Under normoxia, hypoxia inducible factor (HIF) α subunits are hydroxylated by PHDs (prolyl hydroxylase domain proteins) and subsequently undergo polyubiquitylation and degradation. Normal embryogenesis occurs under hypoxia, which suppresses PHD activities and allows HIFα to stabilize and regulate development. In this Primer, we explain molecular mechanisms of the oxygen-sensing pathway, summarize HIF-regulated downstream events, discuss loss-of-function phenotypes primarily in mouse development, and highlight clinical relevance to angiogenesis and tissue repair.
- Published
- 2021
- Full Text
- View/download PDF
16. Isoform-specific Roles of Prolyl Hydroxylases in the Regulation of Pancreatic β-Cell Function
- Author
-
Emelien Jentz, David J. Hodson, Daniela Nasteska, A. Russell Tupling, Federica Cuozzo, Guo-Hua Fong, Monica Hoang, Jamie W. Joseph, and Sarah M. Janssen
- Subjects
Male ,insulin secretion ,medicine.medical_treatment ,Apoptosis ,Oxidative Phosphorylation ,Mice ,Endocrinology ,Adenosine Triphosphate ,cell metabolism ,Insulin-Secreting Cells ,Glucose homeostasis ,Homeostasis ,Insulin ,Protein Isoforms ,insulin release ,HIF1α ,Mice, Knockout ,0303 health sciences ,geography.geographical_feature_category ,islet ,Chemistry ,030302 biochemistry & molecular biology ,Islet ,Adenosine Diphosphate ,Phenotype ,ARNT/HIF1β ,Ketoglutaric Acids ,AcademicSubjects/MED00250 ,Research Article ,medicine.medical_specialty ,Cell signaling ,PHD ,prolyl hydroxylases ,03 medical and health sciences ,Insulin resistance ,Oxygen Consumption ,Protein Domains ,In vivo ,Internal medicine ,medicine ,Animals ,Secretion ,030304 developmental biology ,pancreatic β-cell ,geography ,hypoxia ,Metabolism ,Glucose Tolerance Test ,medicine.disease ,Hypoxia-Inducible Factor 1, alpha Subunit ,Mice, Inbred C57BL ,Glucose ,Gene Expression Regulation ,metabolism ,NADP - Abstract
Pancreatic β-cells can secrete insulin via 2 pathways characterized as KATP channel -dependent and -independent. The KATP channel–independent pathway is characterized by a rise in several potential metabolic signaling molecules, including the NADPH/NADP+ ratio and α-ketoglutarate (αKG). Prolyl hydroxylases (PHDs), which belong to the αKG-dependent dioxygenase superfamily, are known to regulate the stability of hypoxia-inducible factor α. In the current study, we assess the role of PHDs in vivo using the pharmacological inhibitor dimethyloxalylglycine (DMOG) and generated β-cell-specific knockout (KO) mice for all 3 isoforms of PHD (β-PHD1 KO, β-PHD2 KO, and β-PHD3 KO mice). DMOG inhibited in vivo insulin secretion in response to glucose challenge and inhibited the first phase of insulin secretion but enhanced the second phase of insulin secretion in isolated islets. None of the β-PHD KO mice showed any significant in vivo defects associated with glucose tolerance and insulin resistance except for β-PHD2 KO mice which had significantly increased plasma insulin during a glucose challenge. Islets from both β-PHD1 KO and β-PHD3 KO had elevated β-cell apoptosis and reduced β-cell mass. Isolated islets from β-PHD1 KO and β-PHD3 KO had impaired glucose-stimulated insulin secretion and glucose-stimulated increases in the ATP/ADP and NADPH/NADP+ ratio. All 3 PHD isoforms are expressed in β-cells, with PHD3 showing the most distinct expression pattern. The lack of each PHD protein did not significantly impair in vivo glucose homeostasis. However, β-PHD1 KO and β-PHD3 KO mice had defective β-cell mass and islet insulin secretion, suggesting that these mice may be predisposed to developing diabetes.
- Published
- 2021
17. Coupling of angiogenesis and odontogenesis orchestrates tooth mineralization in mice
- Author
-
Tomoko Matsubara, Takahito Iga, Yuki Sugiura, Dai Kusumoto, Tsukasa Sanosaka, Ikue Tai-Nagara, Norihiko Takeda, Guo-Hua Fong, Kosei Ito, Masatsugu Ema, Hideyuki Okano, Jun Kohyama, Makoto Suematsu, and Yoshiaki Kubota
- Subjects
Mice ,Odontoblasts ,Osteogenesis ,Immunology ,Immunology and Allergy ,Animals ,Endothelial Cells ,Odontogenesis ,Cell Differentiation - Abstract
The skeletal system consists of bones and teeth, both of which are hardened via mineralization to support daily physical activity and mastication. The precise mechanism for this process, especially how blood vessels contribute to tissue mineralization, remains incompletely understood. Here, we established an imaging technique to visualize the 3D structure of the tooth vasculature at a single-cell level. Using this technique combined with single-cell RNA sequencing, we identified a unique endothelial subtype specialized to dentinogenesis, a process of tooth mineralization, termed periodontal tip-like endothelial cells. These capillaries exhibit high angiogenic activity and plasticity under the control of odontoblasts; in turn, the capillaries trigger odontoblast maturation. Metabolomic analysis demonstrated that the capillaries perform the phosphate delivery required for dentinogenesis. Taken together, our data identified the fundamental cell-to-cell communications that orchestrate tooth formation, angiogenic–odontogenic coupling, a distinct mechanism compared to the angiogenic–osteogenic coupling in bones. This mechanism contributes to our understanding concerning the functional diversity of organotypic vasculature.
- Published
- 2021
18. Dependence of Retinal Pigment Epithelium Integrity on the NRF2–Heme Oxygenase-1 Axis
- Author
-
Yida, Jiang, Li-Juan, Duan, Jingbo, Pi, Yun-Zheng, Le, and Guo-Hua, Fong
- Subjects
Mice ,Oxidative Stress ,NF-E2-Related Factor 2 ,Animals ,Heme ,Retinal Pigment Epithelium ,General Medicine ,Oxidants ,Heme Oxygenase-1 - Abstract
Tight junctions (TJs) form the structural basis of retinal pigment epithelium (RPE) barrier functions. Although oxidative stress contributes to age-related macular degeneration, it is unclear how RPE TJ integrity is controlled by redox balance. In this study, we investigated the protective roles of nuclear factor erythroid 2-related factor 2 (NRF2), a transcription factor, and heme oxygenase-1 (HO1), a heme-degrading enzyme encoded by the NRF2 target gene HMOX1.ARPE19 cell cultures and mice, including wild-type, Nrf2-/-, and RPE-specific NRF2-deficient mice, were treated with chemicals that impose oxidative stress or impact heme metabolism. In addition, NRF2 and HO1 expression in ARPE19 cells was knocked down by siRNA. TJ integrity was examined by anti-zonula occludens-1 staining of cultured cells or flatmount RPE tissues from mice. RPE barrier functions were evaluated by transepithelium electrical resistance in ARPE19 cells and immunofluorescence staining for albumin or dextran in eye histological sections.TJ structures and RPE barrier functions were compromised due to oxidant exposure and NRF2 deficiency but were rescued by HO1 inducer. Furthermore, treatment with HO1 inhibitor or heme precursor is destructive to TJ structures and RPE barrier properties. Interestingly, both NRF2 and HO1 were upregulated under oxidative stress, probably as an adaptive response to mitigate oxidant-inflicted damages.Our data indicate that the NRF2-HO1 axis protects TJ integrity and RPE barrier functions by driving heme degradation.
- Published
- 2022
- Full Text
- View/download PDF
19. Correction: Ablation of endothelial VEGFR1 improves metabolic dysfunction by inducing adipose tissue browning
- Author
-
Mitsuhiko Abe, Xinsheng Wang, Yihai Cao, Carina Fischer, Kayoko Hosaka, Hideki Iwamoto, Takahiro Seki, Sharon Lim, Patrik Andersson, Guo-Hua Fong, and Yanyan Gao
- Subjects
Pathology ,medicine.medical_specialty ,Endothelium ,business.industry ,medicine.medical_treatment ,Immunology ,Adipose tissue ,Correction ,Ablation ,Article ,medicine.anatomical_structure ,Metabolic disturbance ,medicine ,Browning ,Immunology and Allergy ,business ,Research Articles - Abstract
Seki et al. show that ablation of endothelial VEGFR1 induces adipose tissues browning in healthy and obese mice, which has profound effects on improving global metabolic dysfunctions. These discoveries establish an important role for the adipose vasculature in controlling the metabolic functions of adipocytes and provide new therapeutic options for treatment of obesity and diabetes., Angiogenesis plays an instrumental role in the modulation of adipose tissue mass and metabolism. Targeting adipose vasculature provides an outstanding opportunity for treatment of obesity and metabolic disorders. Here, we report the physiological functions of VEGFR1 in the modulation of adipose angiogenesis, obesity, and global metabolism. Pharmacological inhibition and genetic deletion of endothelial VEGFR1 augmented adipose angiogenesis and browning of subcutaneous white adipose tissue, leading to elevated thermogenesis. In a diet-induced obesity model, endothelial-VEGFR1 deficiency demonstrated a potent anti-obesity effect by improving global metabolism. Along with metabolic changes, fatty liver and insulin sensitivity were also markedly improved in VEGFR1-deficient high fat diet (HFD)–fed mice. Together, our data indicate that targeting of VEGFR1 provides an exciting new opportunity for treatment of obesity and metabolic diseases, such as liver steatosis and type 2 diabetes.
- Published
- 2020
20. Retinal Angiogenesis Regulates Astrocytic Differentiation in Neonatal Mouse Retinas by Oxygen Dependent Mechanisms
- Author
-
Guo-Hua Fong, Thomas N. Sato, Li-Juan Duan, and Sarah J. Pan
- Subjects
0301 basic medicine ,Angiogenesis ,Neurogenesis ,Cellular differentiation ,Optic Disk ,Neovascularization, Physiologic ,lcsh:Medicine ,Hyperoxia ,Retinal Neovascularization ,Article ,Retina ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Basic Helix-Loop-Helix Transcription Factors ,medicine ,Animals ,lcsh:Science ,Multidisciplinary ,Neovascularization, Pathologic ,Chemistry ,Stem Cells ,lcsh:R ,Retinal Vessels ,Cell Differentiation ,Retinal ,Vascular Endothelial Growth Factor Receptor-2 ,Cell biology ,Oxygen ,Endothelial stem cell ,030104 developmental biology ,medicine.anatomical_structure ,Animals, Newborn ,Hypoxia-inducible factors ,Astrocytes ,Optic nerve ,lcsh:Q ,medicine.symptom ,030217 neurology & neurosurgery ,Astrocyte - Abstract
In mice, retinal vascular and astrocyte networks begin to develop at birth, expanding radially from the optic nerve head (ONH) towards the retinal periphery. The retinal vasculature grows towards the periphery ahead of differentiated astrocytes, but behind astrocytic progenitor cells (APCs) and immature astrocytes. Endothelial cell specific Vegfr-2 disruption in newborn mice not only blocked retinal vascular development but also suppressed astrocytic differentiation, reducing the abundance of differentiated astrocytes while causing the accumulation of precursors. By contrast, retinal astrocytic differentiation was accelerated by the exposure of wild-type newborn mice to hyperoxia for 24 hours, or by APC specific deficiency in hypoxia inducible factor (HIF)−2α, an oxygen labile transcription factor. These findings reveal a novel function of the retinal vasculature, and imply that in normal neonatal mice, oxygen from the retinal circulation may promote astrocytic differentiation, in part by triggering oxygen dependent HIF-2α degradation in astrocytic precursors.
- Published
- 2017
- Full Text
- View/download PDF
21. Inhibition of FLT1 ameliorates muscular dystrophy phenotype by increased vasculature in a mouse model of Duchenne muscular dystrophy
- Author
-
Dennis Keefe, Guo-Hua Fong, Zhiwei Zhou, Mayank Verma, Atsushi Asakura, Jennifer Bosco, Yoko Asakura, Serene Josiah, Yuko Shimizu-Motohashi, and James P. Ennen
- Subjects
Male ,Vascular Endothelial Growth Factor A ,Cancer Research ,Heredity ,Muscle Physiology ,Muscle Functions ,Physiology ,Genetic Linkage ,Duchenne muscular dystrophy ,Duchenne Muscular Dystrophy ,QH426-470 ,Muscular Dystrophies ,chemistry.chemical_compound ,Gene Knockout Techniques ,Mice ,0302 clinical medicine ,Animal Cells ,Conditional gene knockout ,Medicine and Health Sciences ,Muscular dystrophy ,Enzyme-Linked Immunoassays ,Receptor ,Musculoskeletal System ,Genetics (clinical) ,0303 health sciences ,biology ,Muscles ,Antibodies, Monoclonal ,Animal Models ,Vascular endothelial growth factor ,medicine.anatomical_structure ,Experimental Organism Systems ,Physiological Parameters ,Neurology ,X-Linked Traits ,Sex Linkage ,Organ Specificity ,Signal transduction ,Anatomy ,Cellular Types ,Dystrophin ,Research Article ,Signal Transduction ,musculoskeletal diseases ,congenital, hereditary, and neonatal diseases and abnormalities ,Mouse Models ,Research and Analysis Methods ,Muscle Fibers ,03 medical and health sciences ,Model Organisms ,medicine ,Genetics ,Animals ,Immunoassays ,Muscle, Skeletal ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,030304 developmental biology ,Clinical Genetics ,Vascular Endothelial Growth Factor Receptor-1 ,Body Weight ,Skeletal muscle ,Biology and Life Sciences ,Endothelial Cells ,Cell Biology ,Skeletal Muscle Fibers ,medicine.disease ,Muscular Dystrophy, Duchenne ,Disease Models, Animal ,chemistry ,Skeletal Muscles ,biology.protein ,Cancer research ,Animal Studies ,Immunologic Techniques ,Mice, Inbred mdx ,Peptides ,030217 neurology & neurosurgery - Abstract
Duchenne muscular dystrophy (DMD) is an X-linked recessive genetic disease in which the dystrophin coding for a membrane stabilizing protein is mutated. Recently, the vasculature has also shown to be perturbed in DMD and DMD model mdx mice. Recent DMD transcriptomics revealed the defects were correlated to a vascular endothelial growth factor (VEGF) signaling pathway. To reveal the relationship between DMD and VEGF signaling, mdx mice were crossed with constitutive (CAGCreERTM:Flt1LoxP/LoxP) and endothelial cell-specific conditional gene knockout mice (Cdh5CreERT2:Flt1LoxP/LoxP) for Flt1 (VEGFR1) which is a decoy receptor for VEGF. Here, we showed that while constitutive deletion of Flt1 is detrimental to the skeletal muscle function, endothelial cell-specific Flt1 deletion resulted in increased vascular density, increased satellite cell number and improvement in the DMD-associated phenotype in the mdx mice. These decreases in pathology, including improved muscle histology and function, were recapitulated in mdx mice given anti-FLT1 peptides or monoclonal antibodies, which blocked VEGF-FLT1 binding. The histological and functional improvement of dystrophic muscle by FLT1 blockade provides a novel pharmacological strategy for the potential treatment of DMD., Author summary Duchenne muscular dystrophy (DMD) is a devastating muscle disease affecting one in 5,000 newborn males, in which the gene encoding the dystrophin protein is mutated. It is a progressive muscle degenerative disease with death by either respiratory insufficiency or cardiac failure in their 20s. Recently, the vasculature has also shown to be perturbed in DMD and DMD model mdx mice with the defects correlated to a vascular endothelial growth factor (VEGF) signaling pathway. To reveal the relationship between DMD and VEGF signaling, mdx mice were crossed with mice carrying mutated a decoy receptor gene (Flt1) for VEGF. Here, we showed that Flt1 deletion resulted in increased vascular density and improvement in the DMD-associated skeletal muscle phenotype in the mdx mice. These decreases in pathology, including improved muscle histology and function, were recapitulated in mdx mice given anti-FLT1 peptides or monoclonal antibodies, which blocked VEGF-FLT1 binding. The histological and functional improvement of dystrophic muscle by FLT1 blockade provides a novel pharmacological strategy for the potential treatment of DMD.
- Published
- 2019
22. Neuron-derived VEGF contributes to cortical and hippocampal development independently of VEGFR1/2-mediated neurotrophism
- Author
-
Keisuke Okabe, Norihiko Takeda, Guo-Hua Fong, Yoshiaki Kubota, Takao Honda, Hugh Fukada, Masatsugu Ema, Tomofumi Ando, Ikue Tai-Nagara, and Kazunori Nakajima
- Subjects
Male ,Vascular Endothelial Growth Factor A ,Angiogenesis ,Neovascularization, Physiologic ,Biology ,Hippocampal formation ,Hippocampus ,Polymerase Chain Reaction ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Neurotrophic factors ,Parietal Lobe ,medicine ,Animals ,Receptor ,Molecular Biology ,Alleles ,030304 developmental biology ,Mice, Knockout ,Neurons ,0303 health sciences ,Vascular Endothelial Growth Factor Receptor-1 ,Endothelial Cells ,Cell Biology ,Vascular Endothelial Growth Factor Receptor-2 ,Cell biology ,Vascular endothelial growth factor ,Mice, Inbred C57BL ,medicine.anatomical_structure ,chemistry ,Mitogen-activated protein kinase ,Mutation ,biology.protein ,Female ,Neuron ,030217 neurology & neurosurgery ,Developmental Biology ,Neurotrophin - Abstract
Vascular endothelial growth factor (VEGF) is a potent mitogen critical for angiogenesis and organogenesis. Deletion or inhibition of VEGF during development not only profoundly suppresses vascular outgrowth, but significantly affects the development and function of various organs. In the brain, VEGF is thought to not only promote vascular growth, but also directly act on neurons as a neurotrophic factor by activating VEGF receptors. In the present study, we demonstrated that deletion of VEGF using hGfap-Cre line, which recombines genes specifically in cortical and hippocampal neurons, severely impaired brain organization and vascularization of these regions. The mutant mice had motor deficits, with lethality around the time of weaning. Multiple reporter lines indicated that VEGF was highly expressed in neurons, but that its cognate receptors, VEGFR1 and 2 were exclusive to endothelial cells in the brain. In accordance, mice lacking neuronal VEGFR1 and VEGFR2 did not exhibit neuronal deformities or lethality. Taken together, our data suggest that neuron-derived VEGF contributes to cortical and hippocampal development likely through angiogenesis independently of direct neurotrophic effects mediated by VEGFR1 and 2.
- Published
- 2019
23. Amelioration of muscular dystrophy phenotype in mdx mice by inhibition of Flt1
- Author
-
Yoko Asakura, Mayank Verma, Jennifer Bosco, Serene Josiah, James P. Ennen, Atsushi Asakura, Yuko Shimizu-Motohashi, Zhiwei Zou, Guo-Hua Fong, and Dennis Keefe
- Subjects
musculoskeletal diseases ,congenital, hereditary, and neonatal diseases and abnormalities ,0303 health sciences ,Duchenne muscular dystrophy ,Skeletal muscle ,Biology ,medicine.disease ,3. Good health ,Vascular endothelial growth factor ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,medicine.anatomical_structure ,chemistry ,Conditional gene knockout ,Knockout mouse ,medicine ,Cancer research ,biology.protein ,Muscular dystrophy ,Receptor ,Dystrophin ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
Duchenne muscular dystrophy (DMD) is an X-linked recessive genetic disease in which the dystrophin coding for a membrane stabilizing protein is mutated. Recently, the vasculature has also shown to be perturbed in DMD and DMD modelmdxmice. Data-mining DMD transcriptomics revealed the defects were correlated to a vascular endothelial growth factor (VEGF) signaling pathway. To reveal the relationship between DMD and VEGF signaling,mdxmice were crossed with constitutive (CAG/CreERTM:Flt1LoxP/LoxP) and endothelial cell-specific conditional gene knockout mice (Cdh5CreERT2:Flt1LoxP/LoxP) forFlt1which is a decoy receptor for VEGF. Previous work demonstrated that heterozygous globalFlt1knockout mice increased vascular density and improved DMD phenotypes when crossed with DMD modelmdxandmdx:utrn-/-mice. Here, we showed that while constitutive deletion ofFlt1is detrimental to the skeletal muscle function, endothelial cell-specificFlt1deletion resulted in increased vascular density and improvement in the DMD-associated phenotype in themdxmice. These decreases in pathology, including improved muscle histology and function, were recapitulated inmdxmice given anti-FLT1 peptides or monoclonal antibodies, which blocked VEGF-FLT1 binding. The histological and functional improvement of dystrophic muscle by FLT1 blockade provides a novel pharmacological strategy for the potential treatment of DMD.
- Published
- 2019
- Full Text
- View/download PDF
24. Assembly of Bifunctional Aptamer–Fibrinogen Macromer for VEGF Delivery and Skin Wound Healing
- Author
-
Xiaolong Zhang, Ming Xu, Akiho Suzuki, Jinping Lai, James Coyne, Yong Wang, Na Xiong, Nan Zhao, Peng Shi, and Guo-Hua Fong
- Subjects
Chemistry ,Angiogenesis ,General Chemical Engineering ,Aptamer ,food and beverages ,02 engineering and technology ,General Chemistry ,010402 general chemistry ,021001 nanoscience & nanotechnology ,01 natural sciences ,Regenerative medicine ,Article ,0104 chemical sciences ,Macromolecular assembly ,Vascular endothelial growth factor ,chemistry.chemical_compound ,Molecular recognition ,Drug delivery ,Self-healing hydrogels ,Materials Chemistry ,Biophysics ,0210 nano-technology - Abstract
Macromolecular assembly has been studied for various applications. However, while macromolecules can recognize one another for assembly, their assembled structures usually lack the function of specific molecular recognition. We hypothesized that bifunctional aptamer-protein macromers would possess dual functions of molecular assembly and recognition. The data show that hybrid aptamer-fibrinogen macromers can assemble to form hydrogels. Moreover, the assembled hydrogels can recognize vascular endothelial growth factor (VEGF) for sustained release. When the VEGF-loaded hydrogels are implanted in vivo, they can promote angiogenesis and skin wound healing. Thus, this work has successfully demonstrated a promising macromolecular system for broad applications such as drug delivery and regenerative medicine.
- Published
- 2019
25. Developmental vascular pruning in neonatal mouse retinas is programmed in the astrocytic oxygen sensing mechanism
- Author
-
Guo-Hua Fong and Li-Juan Duan
- Subjects
Vascular Endothelial Growth Factor A ,Angiogenesis ,Population ,Apoptosis ,Stimulation ,Biology ,Retina ,Hypoxia-Inducible Factor-Proline Dioxygenases ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Cell Movement ,medicine ,Animals ,Pseudopodia ,education ,Molecular Biology ,Oxygen sensing ,Cell Proliferation ,030304 developmental biology ,0303 health sciences ,education.field_of_study ,Neonatal mouse ,Retinal ,Hypoxia (medical) ,Cell biology ,Oxygen ,Hypoxia-inducible factors ,chemistry ,Astrocytes ,medicine.symptom ,030217 neurology & neurosurgery ,Research Article ,Developmental Biology - Abstract
Vascular pruning is crucial for normal development, but its underlying mechanisms are poorly understood. Here, we report that retinal vascular pruning is controlled by the oxygen-sensing mechanism in local astrocytes. Oxygen sensing is mediated by prolyl hydroxylase domain proteins (PHDs), which use O(2) as a substrate to hydroxylate specific prolyl residues on hypoxia inducible factor (HIF)-α proteins, labeling them for polyubiquitylation and proteasomal degradation. In neonatal mice, astrocytic PHD2 deficiency led to elevated HIF-2α protein levels, expanded retinal astrocyte population and defective vascular pruning. Although astrocytic VEGF-A was also increased, anti-VEGF failed to rescue vascular pruning. However, stimulation of retinal astrocytic growth by intravitreal delivery of PDGF-A was sufficient to block retinal vascular pruning in wild-type mice. We propose that in normal development, oxygen from nascent retinal vasculature triggers PHD2-dependent HIF-2α degradation in nearby astrocytic precursors, thus limiting their further growth by driving them to differentiate into non-proliferative mature astrocytes. The physiological limit of retinal capillary density may be set by astrocytes available to support their survival, with excess capillaries destined for regression. This article has an associated ‘The people behind the papers’ interview.
- Published
- 2019
- Full Text
- View/download PDF
26. Erythropoietin Synthesis in Renal Myofibroblasts Is Restored by Activation of Hypoxia Signaling
- Author
-
Sadayoshi Ito, Hiroki Sekine, Akira Nishiyama, Masayuki Yamamoto, Guo-Hua Fong, Takashi Dan, Kotaro Takeda, Masahiro Nezu, Toshio Miyata, Shun Yamazaki, Norio Suzuki, Daisuke Nakano, Tomokazu Souma, and Ikuo Hirano
- Subjects
0301 basic medicine ,Genetically modified mouse ,Endothelium ,Transgene ,Inflammation ,Kidney ,Mice ,03 medical and health sciences ,Fibrosis ,hemic and lymphatic diseases ,medicine ,Renal fibrosis ,Animals ,Myofibroblasts ,Erythropoietin ,business.industry ,General Medicine ,Hypoxia (medical) ,medicine.disease ,Cell Hypoxia ,Basic Research ,030104 developmental biology ,medicine.anatomical_structure ,Nephrology ,Immunology ,Cancer research ,medicine.symptom ,business ,Signal Transduction ,medicine.drug - Abstract
Erythropoietin (Epo) is produced by renal Epo-producing cells (REPs) in a hypoxia-inducible manner. The conversion of REPs into myofibroblasts and coincident loss of Epo-producing ability are the major cause of renal fibrosis and anemia. However, the hypoxic response of these transformed myofibroblasts remains unclear. Here, we used complementary in vivo transgenic and live imaging approaches to better understand the importance of hypoxia signaling in Epo production. Live imaging of REPs in transgenic mice expressing green fluorescent protein from a modified Epo-gene locus revealed that healthy REPs tightly associated with endothelium by wrapping processes around capillaries. However, this association was hampered in states of renal injury-induced inflammation previously shown to correlate with the transition to myofibroblast-transformed renal Epo-producing cells (MF-REPs). Furthermore, activation of hypoxia-inducible factors (HIFs) by genetic inactivation of HIF-prolyl hydroxylases (PHD1, PHD2, and PHD3) selectively in Epo-producing cells reactivated Epo production in MF-REPs. Loss of PHD2 in REPs restored Epo-gene expression in injured kidneys but caused polycythemia. Notably, combined deletions of PHD1 and PHD3 prevented loss of Epo expression without provoking polycythemia. Mice with PHD-deficient REPs also showed resistance to LPS-induced Epo repression in kidneys, suggesting that augmented HIF signaling counterbalances inflammatory stimuli in regulation of Epo production. Thus, augmentation of HIF signaling may be an attractive therapeutic strategy for treating renal anemia by reactivating Epo synthesis in MF-REPs.
- Published
- 2016
- Full Text
- View/download PDF
27. PHD3 Stabilizes the Tight Junction Protein Occludin and Protects Intestinal Epithelial Barrier Function
- Author
-
Yi-Ming Xu, Ying Chen, Guo-Hua Fong, Tang-Long Yuan, Hai-Sheng Zhang, Guohao Wu, Heng Sun, Jing(方靖) Fang, Chen-Guang Bai, Qiulei Xi, Yan-Qing Qin, li(范莉) Fan, and Zhi-Qiang Ling
- Subjects
education ,Procollagen-Proline Dioxygenase ,Mice, Transgenic ,Biology ,Occludin ,digestive system ,Biochemistry ,Inflammatory bowel disease ,Mice ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,Intestinal Mucosa ,Colitis ,Molecular Biology ,health care economics and organizations ,Tight junction ,Cell Biology ,NFKB1 ,medicine.disease ,Intestinal epithelium ,digestive system diseases ,Cell biology ,Ubiquitin ligase ,HEK293 Cells ,Hypoxia-inducible factors ,Immunology ,biology.protein ,Gene Deletion - Abstract
Prolyl hydroxylase domain proteins (PHDs) control cellular adaptation to hypoxia. PHDs are found involved in inflammatory bowel disease (IBD); however, the exact role of PHD3, a member of the PHD family, in IBD remains unknown. We show here that PHD3 plays a critical role in maintaining intestinal epithelial barrier function. We found that genetic ablation of Phd3 in intestinal epithelial cells led to spontaneous colitis in mice. Deletion of PHD3 decreases the level of tight junction protein occludin, leading to a failure of intestinal epithelial barrier function. Further studies indicate that PHD3 stabilizes occludin by preventing the interaction between the E3 ligase Itch and occludin, in a hydroxylase-independent manner. Examination of biopsy of human ulcerative colitis patients indicates that PHD3 is decreased with disease severity, indicating that PHD3 down-regulation is associated with progression of this disease. We show that PHD3 protects intestinal epithelial barrier function and reveal a hydroxylase-independent function of PHD3 in stabilizing occludin. These findings may help open avenues for developing a therapeutic strategy for IBD.
- Published
- 2015
- Full Text
- View/download PDF
28. Lacteal junction zippering protects against diet-induced obesity
- Author
-
Georgia Zarkada, Steffen E. Künzel, Feng Zhang, Carlos Fernández-Hernando, William C. Sessa, Patrick Tso, Anne Eichmann, Roxana Ola, Abhishek Singh, Gerald I. Shulman, Michael Simons, Jinyu Li, Gael Genet, Guo-Hua Fong, Jinah Han, Alexandre Dubrac, Kevin Boyé, Pauline Michon, and Joao Paulo Camporez
- Subjects
0301 basic medicine ,Male ,Vascular Endothelial Growth Factor A ,Lacteal ,Dietary lipid ,Diet, High-Fat ,Article ,03 medical and health sciences ,Mice ,Antigens, CD ,Neuropilin 1 ,Chylomicrons ,medicine ,Animals ,Obesity ,Receptor ,Cytoskeleton ,Mice, Knockout ,Multidisciplinary ,Vascular Endothelial Growth Factor Receptor-1 ,Chemistry ,Cadherins ,Dietary Fats ,Vascular Endothelial Growth Factor Receptor-2 ,Neuropilin-1 ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Lymphatic system ,Enterocytes ,Intestinal Absorption ,Signal transduction ,Gene Deletion ,Chylomicron ,Signal Transduction - Abstract
Zipping up obesity Chylomicrons are specialized particles that carry dietary fats from the intestine to the bloodstream for absorption into the body. Lacteals are lymphatic vessels that act as the highway for chylomicron transport, but it is unclear how passage occurs. Zhang et al. report that two endothelial cell receptors, neuropilin-1 (NRP1) and vascular endothelial growth factor receptor 1 (VEGFR1, also known as FLT1), are required to convert the entry spaces between lacteals from open junctions to closed, zipped structures (see the Perspective by McDonald). Mice that were fed a high-fat diet were subsequently rendered resistant to weight gain if NRP1 and FLT1 were inactivated. Science , this issue p. 599 ; see also p. 551
- Published
- 2017
29. Ablation of endothelial VEGFR1 improves metabolic dysfunction by inducing adipose tissue browning
- Author
-
Guo-Hua Fong, Hideki Iwamoto, Patrik Andersson, Yanyan Gao, Mitsuhiko Abe, Kayoko Hosaka, Sharon Lim, Xinsheng Wang, Yihai Cao, Takahiro Seki, and Carina Fischer
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Endothelium ,Angiogenesis ,Adipose Tissue, White ,Immunology ,Adipose tissue ,Neovascularization, Physiologic ,Type 2 diabetes ,White adipose tissue ,Diet, High-Fat ,Neovascularization ,03 medical and health sciences ,Mice ,Adipose Tissue, Brown ,Metabolic Diseases ,Internal medicine ,medicine ,Immunology and Allergy ,Animals ,Obesity ,Mice, Knockout ,Vascular Endothelial Growth Factor Receptor-1 ,business.industry ,Fatty liver ,Thermogenesis ,medicine.disease ,3. Good health ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,Adipose Tissue ,Female ,Endothelium, Vascular ,medicine.symptom ,business - Abstract
Angiogenesis plays an instrumental role in the modulation of adipose tissue mass and metabolism. Targeting adipose vasculature provides an outstanding opportunity for treatment of obesity and metabolic disorders. Here, we report the physiological functions of VEGFR1 in the modulation of adipose angiogenesis, obesity, and global metabolism. Pharmacological inhibition and genetic deletion of endothelial VEGFR1 augmented adipose angiogenesis and browning of subcutaneous white adipose tissue, leading to elevated thermogenesis. In a diet-induced obesity model, endothelial-VEGFR1 deficiency demonstrated a potent anti-obesity effect by improving global metabolism. Along with metabolic changes, fatty liver and insulin sensitivity were also markedly improved in VEGFR1-deficient high fat diet (HFD)–fed mice. Together, our data indicate that targeting of VEGFR1 provides an exciting new opportunity for treatment of obesity and metabolic diseases, such as liver steatosis and type 2 diabetes.
- Published
- 2017
30. PTEN induces apoptosis and cavitation via HIF-2-dependent Bnip3 upregulation during epithelial lumen formation
- Author
-
J Liu, Saadat S, Han Y, Lee Ly, Pier Paolo Pandolfi, Guo-Hua Fong, Li S, Qi Y, and Tian X
- Subjects
Mammalian embryology ,Apoptosis ,Embryoid body ,Epithelium ,Mitochondrial Proteins ,Mice ,Downregulation and upregulation ,Animals ,PTEN ,Tensin ,Molecular Biology ,Protein kinase B ,Mice, Knockout ,Regulation of gene expression ,Original Paper ,biology ,PTEN Phosphohydrolase ,Gene Expression Regulation, Developmental ,Membrane Proteins ,Cell Biology ,Embryo, Mammalian ,Up-Regulation ,Cell biology ,Cancer research ,biology.protein ,Proto-Oncogene Proteins c-akt ,Transcription Factors - Abstract
The tumor suppressor phosphatase and tensin homolog (PTEN) dephosphorylates PIP3 and antagonizes the prosurvival PI3K-Akt pathway. Targeted deletion of PTEN in mice led to early embryonic lethality. To elucidate its role in embryonic epithelial morphogenesis and the underlying mechanisms, we used embryonic stem cell-derived embryoid body (EB), an epithelial cyst structurally similar to the periimplantation embryo. PTEN is upregulated during EB morphogenesis in parallel with apoptosis of core cells, which mediates EB cavitation. Genetic ablation of PTEN causes Akt overactivation, apoptosis resistance and cavitation blockade. However, rescue experiments using mutant PTEN and pharmacological inhibition of Akt suggest that the phosphatase activity of PTEN and Akt are not involved in apoptosis-mediated cavitation. Instead, hypoxia-induced upregulation of Bnip3, a proapoptotic BH3-only protein, mediates PTEN-dependent apoptosis and cavitation. PTEN inactivation inhibits hypoxia- and reactive oxygen species-induced Bnip3 elevation. Overexpression of Bnip3 in PTEN-null EBs rescues apoptosis of the core cells. Mechanistically, suppression of Bnip3 following PTEN loss is likely due to reduction of hypoxia-inducible factor-2α (HIF-2α) because forced expression of an oxygen-stable HIF-2α mutant rescues Bnip3 expression and apoptosis. Lastly, we show that HIF-2α is upregulated by PTEN at both transcriptional and posttranscriptional levels. Ablation of prolyl hydroxylase domain-containing protein 2 (PHD2) in normal EBs or inhibition of PHD activities in PTEN-null EBs stabilizes HIF-2α and induces Bnip3 and caspase-3 activation. Altogether, these results suggest that PTEN is required for apoptosis-mediated cavitation during epithelial morphogenesis by regulating the expression of HIF-2α and Bnip3.
- Published
- 2014
- Full Text
- View/download PDF
31. Broad Suppression of NADPH Oxidase Activity Exacerbates Ischemia/Reperfusion Injury Through Inadvertent Downregulation of Hypoxia-inducible Factor-1α and Upregulation of Peroxisome Proliferator–activated Receptor-α
- Author
-
Yoshiyuki Ikeda, Tetsuro Ago, Guo-Hua Fong, Junya Kuroda, Peiyong Zhai, Shinichi Oka, Rong Tian, Junichi Sadoshima, and Shouji Matsushima
- Subjects
chemistry.chemical_classification ,Genetically modified mouse ,Reactive oxygen species ,medicine.medical_specialty ,NADPH oxidase ,biology ,Physiology ,Peroxisome proliferator-activated receptor ,NOX4 ,medicine.disease_cause ,medicine.disease ,Endocrinology ,chemistry ,Downregulation and upregulation ,Internal medicine ,Immunology ,cardiovascular system ,biology.protein ,medicine ,Cardiology and Cardiovascular Medicine ,Reperfusion injury ,Oxidative stress - Abstract
Rationale: NADPH oxidase (Nox) 2 and Nox4 are major components of the Nox family which purposefully produce reactive oxidative species, namely O 2 − and H 2 O 2 , in the heart. The isoform-specific contribution of Nox2 and Nox4 to ischemia/reperfusion (I/R) injury is poorly understood. Objective: We investigated the role of Nox2 and Nox4 in mediating oxidative stress and myocardial injury during I/R using loss-of-function mouse models. Methods and Results: Systemic (s) Nox2 knockout (KO), sNox4 KO, and cardiac-specific (c) Nox4 KO mice were subjected to I/R (30 minutes/24 hours, respectively). Both myocardial infarct size/area at risk and O 2 − production were lower in sNox2 KO, sNox4 KO, and cNox4 KO than in wild-type mice. Unexpectedly, however, the myocardial infarct size/area at risk was greater, despite less O 2 − production, in sNox2 KO+cNox4 KO (double-KO) mice and transgenic mice (Tg) with cardiac-specific expression of dominant-negative Nox, which suppresses both Nox2 and Nox4, than in wild-type or single KO mice. Hypoxia-inducible factor-1α was downregulated whereas peroxisome proliferator–activated receptor-α was upregulated in Tg-dominant-negative Nox mice. A cross with mice deficient in prolyl hydroxylase 2, which hydroxylates hypoxia-inducible factor-1α, rescued the I/R injury and prevented upregulation of peroxisome proliferator–activated receptor-α in Tg-dominant–negative Nox mice. A cross with peroxisome proliferator–activated receptor-α KO mice also attenuated the injury in Tg- dominant–negative Nox mice. Conclusions: Both Nox2 and Nox4 contribute to the increase in reactive oxidative species and injury by I/R. However, low levels of reactive oxidative species produced by either Nox2 or Nox4 regulate hypoxia-inducible factor-1α and peroxisome proliferator–activated receptor-α, thereby protecting the heart against I/R, suggesting that Noxs also act as a physiological sensor for myocardial adaptation.
- Published
- 2013
- Full Text
- View/download PDF
32. Distinct subpopulations of FOXD1 stroma-derived cells regulate renal erythropoietin
- Author
-
Annika Wernerson, Guo-Hua Fong, Agnes B. Fogo, Qingdu Liu, Andrew S. Pfaff, Andres A. Urrutia, Thomas C. Binns, Kenneth W. Gross, Pinelopi P. Kapitsinou, Volker H. Haase, Olena Davidoff, Hannes Olauson, and Hanako Kobayashi
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Stromal cell ,Cell ,030232 urology & nephrology ,Procollagen-Proline Dioxygenase ,Biology ,urologic and male genital diseases ,Kidney ,Hypoxia-Inducible Factor-Proline Dioxygenases ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Stroma ,Internal medicine ,hemic and lymphatic diseases ,medicine ,Basic Helix-Loop-Helix Transcription Factors ,Animals ,Hypoxia ,Erythropoietin ,Renal stem cell ,Mice, Knockout ,Kidney metabolism ,Forkhead Transcription Factors ,General Medicine ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Cancer research ,Erythropoiesis ,Stromal Cells ,medicine.drug ,Research Article - Abstract
Renal peritubular interstitial fibroblast-like cells are critical for adult erythropoiesis, as they are the main source of erythropoietin (EPO). Hypoxia-inducible factor 2 (HIF-2) controls EPO synthesis in the kidney and liver and is regulated by prolyl-4-hydroxylase domain (PHD) dioxygenases PHD1, PHD2, and PHD3, which function as cellular oxygen sensors. Renal interstitial cells with EPO-producing capacity are poorly characterized, and the role of the PHD/HIF-2 axis in renal EPO-producing cell (REPC) plasticity is unclear. Here we targeted the PHD/HIF-2/EPO axis in FOXD1 stroma-derived renal interstitial cells and examined the role of individual PHDs in REPC pool size regulation and renal EPO output. Renal interstitial cells with EPO-producing capacity were entirely derived from FOXD1-expressing stroma, and Phd2 inactivation alone induced renal Epo in a limited number of renal interstitial cells. EPO induction was submaximal, as hypoxia or pharmacologic PHD inhibition further increased the REPC fraction among Phd2-/- renal interstitial cells. Moreover, Phd1 and Phd3 were differentially expressed in renal interstitium, and heterozygous deficiency for Phd1 and Phd3 increased REPC numbers in Phd2-/- mice. We propose that FOXD1 lineage renal interstitial cells consist of distinct subpopulations that differ in their responsiveness to Phd2 inactivation and thus regulation of HIF-2 activity and EPO production under hypoxia or conditions of pharmacologic or genetic PHD inactivation.
- Published
- 2016
33. Therapeutic targeting of oxygen-sensing prolyl hydroxylases abrogates ATF4-dependent neuronal death and improves outcomes after brain hemorrhage in several rodent models
- Author
-
Frederick Colbourne, Guo Li Ming, David W. Killilea, Giovanni Coppola, John W. Cave, Theodore R. Holman, Hongjun Song, Roseleen F. John, Irina G. Gazaryan, Guohua Xi, Sama F. Sleiman, Frederick R. Maxfield, Guo-Hua Fong, Dana Cruz, Carsten Culmsee, Ishraq Alim, Marina Demetriades, Soah J. Khim, Richard F. Keep, Timothy J Schallert, Cyrille C. Thinnes, Lewis B. Morgenstern, Ryan Tappero, Christopher J. Schofield, Tzu Lan Yeh, Sandra Neitemeier, Megan W. Bourassa, Saravanan S. Karuppagounder, Jian Zhong, Yijing Su, Sunghee Cho, and Rajiv R. Ratan
- Subjects
0301 basic medicine ,Iron ,Procollagen-Proline Dioxygenase ,Oxidative phosphorylation ,Pharmacology ,Iron Chelating Agents ,medicine.disease_cause ,Article ,Toxicology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Protein Domains ,Genes, Reporter ,In vivo ,Animals ,Protein Isoforms ,Medicine ,Molecular Targeted Therapy ,cardiovascular diseases ,Cells, Cultured ,Neurons ,Intracerebral hemorrhage ,chemistry.chemical_classification ,Cell Death ,business.industry ,ATF4 ,Brain ,Recovery of Function ,General Medicine ,Hypoxia-Inducible Factor 1, alpha Subunit ,medicine.disease ,Activating Transcription Factor 4 ,In vitro ,Rats ,Oxygen ,Disease Models, Animal ,Neuroprotective Agents ,030104 developmental biology ,Enzyme ,Gene Expression Regulation ,chemistry ,Hemin ,Liberation ,business ,Intracranial Hemorrhages ,030217 neurology & neurosurgery ,Oxidative stress - Abstract
Disability or death due to intracerebral hemorrhage (ICH) is attributed to blood lysis, liberation of iron, and consequent oxidative stress. Iron chelators bind to free iron and prevent neuronal death induced by oxidative stress and disability due to ICH, but the mechanisms for this effect remain unclear. We show that the hypoxia-inducible factor prolyl hydroxylase domain (HIF-PHD) family of iron-dependent, oxygen-sensing enzymes are effectors of iron chelation. Molecular reduction of the three HIF-PHD enzyme isoforms in the mouse striatum improved functional recovery after ICH. A low-molecular-weight hydroxyquinoline inhibitor of the HIF-PHD enzymes, adaptaquin, reduced neuronal death and behavioral deficits after ICH in several rodent models without affecting total iron or zinc distribution in the brain. Unexpectedly, protection from oxidative death in vitro or from ICH in vivo by adaptaquin was associated with suppression of activity of the prodeath factor ATF4 rather than activation of an HIF-dependent prosurvival pathway. Together, these findings demonstrate that brain-specific inactivation of the HIF-PHD metalloenzymes with the blood-brain barrier–permeable inhibitor adaptaquin can improve functional outcomes after ICH in several rodent models.
- Published
- 2016
34. Disruption of Hypoxia-Inducible Transcription Factor-Prolyl Hydroxylase Domain-1 (PHD-1−/−) Attenuates Ex Vivo Myocardial Ischemia/Reperfusion Injury Through Hypoxia-Inducible Factor-1α Transcription Factor and Its Target Genes in Mice
- Author
-
Juan A. Sanchez, Gautam Maulik, Babatunde Oriowo, Guo-Hua Fong, Nageswara Rao Dunna, Ram Sudheer Adluri, Kotaro Takeda, Mahesh Thirunavukkarasu, Hajime Otani, Nilanjana Maulik, and Lijun Zhan
- Subjects
Nitric Oxide Synthase Type III ,Physiology ,Heart Ventricles ,education ,Clinical Biochemistry ,Myocardial Ischemia ,Procollagen-Proline Dioxygenase ,Ischemia ,Gene Expression ,Apoptosis ,Electrophoretic Mobility Shift Assay ,Myocardial Reperfusion Injury ,In Vitro Techniques ,Biology ,Biochemistry ,Ventricular Function, Left ,Gene Knockout Techniques ,Mice ,hemic and lymphatic diseases ,medicine ,Animals ,Molecular Biology ,Transcription factor ,beta Catenin ,health care economics and organizations ,General Environmental Science ,Cell Nucleus ,Mice, Knockout ,Cardioprotection ,Forum Original Research Communications ,NF-kappa B ,Cell Biology ,Hypoxia (medical) ,Hypoxia-Inducible Factor 1, alpha Subunit ,medicine.disease ,Molecular biology ,Protein Structure, Tertiary ,Protein Transport ,Proto-Oncogene Proteins c-bcl-2 ,Hypoxia-inducible factors ,General Earth and Planetary Sciences ,medicine.symptom ,Reperfusion injury ,Ex vivo ,Protein Binding - Abstract
Hypoxia-inducible transcription factor (HIF)-prolyl hydroxylases domain (PHD-1–3) are oxygen sensors that regulate the stability of the HIFs in an oxygen-dependent manner. Suppression of PHD enzymes leads to stabilization of HIFs and offers a potential treatment option for many ischemic disorders, such as peripheral artery occlusive disease, myocardial infarction, and stroke. Here, we show that homozygous disruption of PHD-1 (PHD-1−/−) could facilitate HIF-1α-mediated cardioprotection in ischemia/reperfused (I/R) myocardium. Wild-type (WT) and PHD-1−/− mice were randomized into WT time-matched control (TMC), PHD-1−/− TMC (PHD1TMC), WT I/R, and PHD-1−/− I/R (PHD1IR). Isolated hearts from each group were subjected to 30 min of global ischemia followed by 2 h of reperfusion. TMC hearts were perfused for 2 h 30 min without ischemia. Decreased infarct size (35% ± 0.6% vs. 49% ± 0.4%) and apoptotic cardiomyocytes (106 ± 13 vs. 233 ± 21 counts/100 high-power field) were observed in PHD1IR compared to wild-type ischemia/reperfusion (WTIR). Protein expression of HIF-1α was significantly increased in PHD1IR compared to WTIR. mRNA expression of β-catenin (1.9-fold), endothelial nitric oxide synthase (1.9-fold), p65 (1.9-fold), and Bcl-2 (2.7-fold) were upregulated in the PHD1IR compared with WTIR, which was studied by real-time quantitative polymerase chain reaction. Further, gel-shift analysis showed increased DNA binding activity of HIF-1α and nuclear factor-kappaB in PHD1IR compared to WTIR. In addition, nuclear translocation of β-catenin was increased in PHD1IR compared with WTIR. These findings indicated that silencing of PHD-1 attenuates myocardial I/R injury probably by enhancing HIF-1α/β-catenin/endothelial nitric oxide synthase/nuclear factor-kappaB and Bcl-2 signaling pathway. Antioxid. Redox Signal. 15, 1789–1797.
- Published
- 2011
- Full Text
- View/download PDF
35. Flt-1 haploinsufficiency ameliorates muscular dystrophy phenotype by developmentally increased vasculature in mdx mice
- Author
-
Christopher Tastad, Guo-Hua Fong, Dawn A. Lowe, Mayank Verma, Jarrod A. Call, Shuichi Watanabe, Masatsugu Ema, Atsushi Asakura, Yoko Asakura, and Hiroyuki Hirai
- Subjects
musculoskeletal diseases ,Heterozygote ,congenital, hereditary, and neonatal diseases and abnormalities ,medicine.medical_specialty ,Membrane permeability ,Duchenne muscular dystrophy ,Haploinsufficiency ,Mice ,chemistry.chemical_compound ,Internal medicine ,Utrophin ,Genetics ,medicine ,Animals ,Muscular dystrophy ,Muscle, Skeletal ,Molecular Biology ,Genetics (clinical) ,Gene knockout ,Cell Proliferation ,Mice, Knockout ,Vascular Endothelial Growth Factor Receptor-1 ,biology ,Homozygote ,Skeletal muscle ,Articles ,General Medicine ,Muscular Dystrophy, Animal ,musculoskeletal system ,medicine.disease ,Vascular endothelial growth factor ,Phenotype ,medicine.anatomical_structure ,Endocrinology ,chemistry ,Immunology ,Mice, Inbred mdx ,cardiovascular system ,biology.protein ,sense organs ,Dystrophin ,Muscle Contraction - Abstract
Duchenne muscular dystrophy (DMD) is an X-linked recessive genetic disease caused by mutations in the gene coding for the protein dystrophin. Recent work demonstrates that dystrophin is also found in the vasculature and its absence results in vascular deficiency and abnormal blood flow. This induces a state of ischemia further aggravating the muscular dystrophy pathogenesis. For an effective form of therapy of DMD, both the muscle and the vasculature need to be addressed. To reveal the developmental relationship between muscular dystrophy and vasculature, mdx mice, an animal model for DMD, were crossed with Flt-1 gene knockout mice to create a model with increased vasculature. Flt-1 is a decoy receptor for vascular endothelial growth factor, and therefore both homozygous (Flt-1(-/-)) and heterozygous (Flt-1(+/-)) Flt-1 gene knockout mice display increased endothelial cell proliferation and vascular density during embryogenesis. Here, we show that Flt-1(+/-) and mdx:Flt-1(+/-) adult mice also display a developmentally increased vascular density in skeletal muscle compared with the wild-type and mdx mice, respectively. The mdx:Flt-1(+/-) mice show improved muscle histology compared with the mdx mice with decreased fibrosis, calcification and membrane permeability. Functionally, the mdx:Flt-1(+/-) mice have an increase in muscle blood flow and force production, compared with the mdx mice. Consequently, the mdx:utrophin(-/-):Flt-1(+/-) mice display improved muscle histology and significantly higher survival rates compared with the mdx:utrophin(-/-) mice, which show more severe muscle phenotypes than the mdx mice. These data suggest that increasing the vasculature in DMD may ameliorate the histological and functional phenotypes associated with this disease.
- Published
- 2010
- Full Text
- View/download PDF
36. Impaired pancreatic development in Hif2-alpha deficient mice
- Author
-
Sanjay K. Mishra, Golbahar Houshmand, Huiping Chen, Farzad Esni, Guo-Hua Fong, and George K. Gittes
- Subjects
Cell signaling ,medicine.medical_specialty ,Organogenesis ,Cellular differentiation ,Biophysics ,Notch signaling pathway ,Biology ,Biochemistry ,Mice ,Precursor cell ,Internal medicine ,Basic Helix-Loop-Helix Transcription Factors ,medicine ,Animals ,Progenitor cell ,Hypoxia ,Pancreas ,Molecular Biology ,Mice, Knockout ,Receptors, Notch ,Cell Biology ,Cell biology ,medicine.anatomical_structure ,Endocrinology ,Notch proteins ,Hes3 signaling axis ,Signal Transduction - Abstract
Accumulating data suggest the existence of a link between hypoxia and maintenance of the undifferentiated cell state, but little is known about the cellular signaling mechanisms underlying this process. Recent reports reveal a direct link between components of the hypoxia signaling pathway and Notch pathway in maintaining precursor cells in an undifferentiated state. Here, we report that in the developing mouse pancreas, Hif2-alpha is expressed in pancreatic progenitor cells, but its expression is lost in committed endocrine progenitors as well as in differentiated endocrine and exocrine cells. In an attempt to analyze the function of HIF2-alpha in the developing pancreas, we studied Hif2-alpha(-/-) pancreas. Our analyses revealed that in addition to the decreased size and branching, the Hif2-alpha deficient pancreas also displayed impaired notch signaling and cell differentiation. Finally, we found that HIF2-alpha binds directly to Notch-IC and that the responsible site for this interaction is within the RAM domain of Notch protein. These results suggest that HIF2-alpha is required for normal mouse pancreatic development.
- Published
- 2010
- Full Text
- View/download PDF
37. Integrity of the prolyl hydroxylase domain protein 2:erythropoietin pathway in aging mice
- Author
-
Guo-Hua Fong, Kotaro Takeda, Scott Sutherland, Xiping Li, and Frank S. Lee
- Subjects
Aging ,Procollagen-Proline Dioxygenase ,Biology ,Article ,Hypoxia-Inducible Factor-Proline Dioxygenases ,Mice ,Downregulation and upregulation ,hemic and lymphatic diseases ,Conditional gene knockout ,medicine ,Animals ,RNA, Messenger ,Erythropoietin ,Molecular Biology ,Transcription factor ,Mice, Knockout ,Regulation of gene expression ,Cell Biology ,Hematology ,Hematopoiesis ,Mice, Inbred C57BL ,Haematopoiesis ,Gene Expression Regulation ,Hypoxia-inducible factors ,Cancer research ,Molecular Medicine ,Hypoxia-Inducible Factor 1 ,Procollagen-proline dioxygenase ,Gene Deletion ,Signal Transduction ,medicine.drug - Abstract
The central transcriptional response to hypoxia is mediated by the prolyl hydroxylase domain protein (PHD):hypoxia inducible factor (HIF) pathway. In this pathway, PHD prolyl hydroxylates and thereby negatively regulates the alpha-subunit of the transcription factor HIF (HIF-alpha). An important HIF target gene is that for erythropoietin (EPO), which controls red cell mass. Recent studies have identified PHD2 as the critical PHD isoform regulating the EPO gene. Other studies have shown that the inducibility of the HIF pathway diminishes as a function of age. Thus, an important question is whether the PHD2:EPO pathway is altered in the aging. Here, we employed a mouse line with a globally-inducible Phd2 conditional knockout allele to examine the integrity of the Phd2:Epo axis in young (six to eight months old) and aging (sixteen to twenty months old) mice. We find that acute global deletion of Phd2 results in a robust erythrocytosis in both young and aging mice, with both age groups showing marked extramedullary hematopoiesis in the spleen. Epo mRNA is dramatically upregulated in the kidney, but not in the liver, in both age groups. Conversely, other Hif targets, including Vegf, Pgk1, and Phd3 are upregulated in the liver but not in the kidney in both age groups. These findings have implications for targeting this pathway in the aging.
- Published
- 2010
- Full Text
- View/download PDF
38. Vascular Endothelial Growth Factor Receptor-1 Regulates Postnatal Angiogenesis Through Inhibition of the Excessive Activation of Akt
- Author
-
Kaoru Tateno, Sho Okada, Junji Moriya, Kenji Sunagawa, Masabumi Shibuya, Aika Nojima, Masayuki Orimo, Jun-ichiro Nishi, Hideyuki Miyauchi, Issei Komuro, Tohru Minamino, and Guo-Hua Fong
- Subjects
Vascular Endothelial Growth Factor A ,Cell Survival ,Physiology ,Angiogenesis ,Neovascularization, Physiologic ,Biology ,Neovascularization ,Mice ,chemistry.chemical_compound ,Ischemia ,medicine ,Animals ,Humans ,Cells, Cultured ,Cellular Senescence ,S1PR1 ,Mice, Knockout ,Vascular Endothelial Growth Factor Receptor-1 ,integumentary system ,Vascular Endothelial Growth Factor Receptor-2 ,Vascular endothelial growth factor ,Endothelial stem cell ,Vascular endothelial growth factor B ,Vascular endothelial growth factor A ,chemistry ,Vascular endothelial growth factor C ,embryonic structures ,cardiovascular system ,Cancer research ,Endothelium, Vascular ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
Vascular endothelial growth factor (VEGF) binds both VEGF receptor-1 (VEGFR-1) and VEGF receptor-2 (VEGFR-2). Activation of VEGFR-2 is thought to play a major role in the regulation of endothelial function by VEGF. Recently, specific ligands for VEGFR-1 have been reported to have beneficial effects when used to treat ischemic diseases. However, the role of VEGFR-1 in angiogenesis is not fully understood. In this study, we showed that VEGFR-1 performs “fine tuning” of VEGF signaling to induce neovascularization. We examined the effects of retroviral vectors expressing a small interference RNA that targeted either the VEGFR-1 gene or the VEGFR-2 gene. Deletion of either VEGFR-1 or VEGFR-2 reduced the ability of endothelial cells to form capillaries. Deletion of VEGFR-1 markedly reduced endothelial cell proliferation and induced premature senescence of endothelial cells. In contrast, deletion of VEGFR-2 significantly impaired endothelial cell survival. When VEGFR-1 expression was blocked, VEGF constitutively activated Akt signals and thus induced endothelial cell senescence via a p53-dependent pathway. VEGFR-1 +/− mice exhibited an increase of endothelial Akt activity and showed an impaired neovascularization in response to ischemia, and this impairment was ameliorated in VEGFR-1 +/− Akt1 +/− mice. These results suggest that VEGFR-1 plays a critical role in the maintenance of endothelial integrity by modulating the VEGF/Akt signaling pathway.
- Published
- 2008
- Full Text
- View/download PDF
39. VEGFR1 Tyrosine Kinase Signaling Promotes Lymphangiogenesis as Well as Angiogenesis Indirectly via Macrophage Recruitment
- Author
-
Yohei Morita, Guo-Hua Fong, Jun Ooehara, Hideo Ema, Toshio Suda, Yujuan Zheng, Masato Murakami, Masabumi Shibuya, and Masanori Hirashima
- Subjects
Male ,Vascular Endothelial Growth Factor A ,Angiogenesis ,Transgene ,Vascular Endothelial Growth Factor C ,Neovascularization, Physiologic ,Mice, Transgenic ,Capillary Permeability ,Neovascularization ,Mice ,medicine ,Animals ,Macrophage ,Lymphangiogenesis ,Receptor ,Bone Marrow Transplantation ,Mice, Knockout ,Mice, Inbred BALB C ,Vascular Endothelial Growth Factor Receptor-1 ,Chemistry ,Macrophages ,Mice, Inbred C57BL ,Immunology ,Cancer research ,Female ,medicine.symptom ,Signal transduction ,Cardiology and Cardiovascular Medicine ,Tyrosine kinase ,Signal Transduction - Abstract
Objective— Angiogenesis and lymphangiogenesis are complex phenomena that involve the interplay of several growth factors and receptors. Recently, we have demonstrated that in Keratin-14 ( K14 ) promoter-driven Vegf-A transgenic (Tg) mice, not only angiogenesis but also lymphangiogenesis is stimulated. However, the mechanism by which VEGFR1 is involved in lymphangiogenesis remains unclear. Methods and Results— To examine how important the tyrosine kinase (TK) of VEGFR1 is in lymphangiogenesis in K14 Vegf-A Tg mice, we crossed the K14 Vegf-A Tg mice with VEGFR1-TK–deficient mice to generate double mutant K14 Vegf-A Tg Vegfr1 tk −/− mice. K14 Vegf-A Tg Vegfr1 tk −/− mice exhibit a remarkable decrease in lymphangiogensis as well as angiogenesis in subcutaneous tissues. To address the mechanism underlying the decrease in lymphangiogensis, we investigated the recruitment of monocyte-macrophage-lineage cells into the skin. The recruitment of VEGFR1-expressing macrophages driven by VEGF-A was reduced in K14 Vegf-A Tg Vegfr1 tk −/− mice. Vegf-A Tg mice that received VEGFR1-TK–deficient bone marrow showed a reduction of macrophage recruitment, lymphangiogenesis and angiogenesis compared with those in K14 Vegf-A Tg mice. Conclusions— VEGFR1 signaling promotes lymphangiogenesis as well as angiogenesis mainly by increasing bone marrow–derived macrophage recruitment.
- Published
- 2008
- Full Text
- View/download PDF
40. Regulation of adult erythropoiesis by prolyl hydroxylase domain proteins
- Author
-
Frank S. Lee, Hiromi Takeda, Guo-Hua Fong, Katie Lamothe, Kotaro Takeda, Nehal S. Parikh, Xiping Li, Hector L. Aguila, and Li-Juan Duan
- Subjects
Aging ,medicine.medical_specialty ,Red Cells ,Immunology ,Procollagen-Proline Dioxygenase ,Biology ,Biochemistry ,Immediate early protein ,Hypoxia-Inducible Factor-Proline Dioxygenases ,Immediate-Early Proteins ,Hydroxylation ,Mice ,chemistry.chemical_compound ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Animals ,Erythropoiesis ,Mice, Knockout ,Kidney ,Cell Biology ,Hematology ,Hematopoietic Stem Cells ,DNA-Binding Proteins ,medicine.anatomical_structure ,Endocrinology ,chemistry ,Erythropoietin ,Procollagen-proline dioxygenase ,Homeostasis ,Intracellular ,medicine.drug - Abstract
Polycythemia is often associated with erythropoietin (EPO) overexpression and defective oxygen sensing. In normal cells, intracellular oxygen concentrations are directly sensed by prolyl hydroxylase domain (PHD)–containing proteins, which tag hypoxia-inducible factor (HIF) α subunits for polyubiquitination and proteasomal degradation by oxygen-dependent prolyl hydroxylation. Here we show that different PHD isoforms differentially regulate HIF-α stability in the adult liver and kidney and suppress Epo expression and erythropoiesis through distinct mechanisms. Although Phd1−/− or Phd3−/− mice had no apparent defects, double knockout of Phd1 and Phd3 led to moderate erythrocytosis. HIF-2α, which is known to activate Epo expression, accumulated in the liver. In adult mice deficient for PHD2, the prototypic Epo transcriptional activator HIF-1α accumulated in both the kidney and liver. Elevated HIF-1α levels were associated with dramatically increased concentrations of both Epo mRNA in the kidney and Epo protein in the serum, which led to severe erythrocytosis. In contrast, heterozygous mutation of Phd2 had no detectable effects on blood homeostasis. These findings suggest that PHD1/3 double deficiency leads to erythrocytosis partly by activating the hepatic HIF-2α/Epo pathway, whereas PHD2 deficiency leads to erythrocytosis by activating the renal Epo pathway.
- Published
- 2008
- Full Text
- View/download PDF
41. Role and regulation of prolyl hydroxylase domain proteins
- Author
-
Guo-Hua Fong and Kotaro Takeda
- Subjects
Gene isoform ,Hypoxia-Inducible Factor 1 ,education ,Procollagen-Proline Dioxygenase ,Biology ,Hydroxylation ,Mice ,chemistry.chemical_compound ,Downregulation and upregulation ,Ubiquitin ,hemic and lymphatic diseases ,Oxygen homeostasis ,Basic Helix-Loop-Helix Transcription Factors ,Animals ,Humans ,Hypoxia ,Molecular Biology ,health care economics and organizations ,Feedback, Physiological ,Cell Biology ,Hypoxia-Inducible Factor 1, alpha Subunit ,Cell Hypoxia ,Cell biology ,Oxygen ,chemistry ,Biochemistry ,biology.protein ,Procollagen-proline dioxygenase ,Signal transduction ,Protein Processing, Post-Translational ,Signal Transduction - Abstract
Oxygen-dependent hydroxylation of hypoxia-inducible factor (HIF)-alpha subunits by prolyl hydroxylase domain (PHD) proteins signals their polyubiquitination and proteasomal degradation, and plays a critical role in regulating HIF abundance and oxygen homeostasis. While oxygen concentration plays a major role in determining the efficiency of PHD-catalyzed hydroxylation reactions, many other environmental and intracellular factors also significantly modulate PHD activities. In addition, PHDs may also employ hydroxylase-independent mechanisms to modify HIF activity. Interestingly, while PHDs regulate HIF-alpha protein stability, PHD2 and PHD3 themselves are subject to feedback upregulation by HIFs. Functionally, different PHD isoforms may differentially contribute to specific pathophysiological processes, including angiogenesis, erythropoiesis, tumorigenesis, and cell growth, differentiation and survival. Because of diverse roles of PHDs in many different processes, loss of PHD expression or function triggers multi-faceted pathophysiological changes as has been shown in mice lacking different PHD isoforms. Future investigations are needed to explore in vivo specificity of PHDs over different HIF-alpha subunits and differential roles of PHD isoforms in different biological processes.
- Published
- 2008
- Full Text
- View/download PDF
42. Essential Role for Prolyl Hydroxylase Domain Protein 2 in Oxygen Homeostasis of the Adult Vascular System
- Author
-
Anne E. Cowan, Guo-Hua Fong, and Kotaro Takeda
- Subjects
Vascular Endothelial Growth Factor A ,Gene isoform ,medicine.medical_specialty ,Vascular smooth muscle ,Angiogenesis ,Procollagen-Proline Dioxygenase ,Neovascularization, Physiologic ,Vasodilation ,Biology ,Hypoxia-Inducible Factor-Proline Dioxygenases ,Immediate-Early Proteins ,Neovascularization ,Mice ,Physiology (medical) ,Internal medicine ,Conditional gene knockout ,Oxygen homeostasis ,Angiopoietin-1 ,medicine ,Animals ,Homeostasis ,Protein Isoforms ,Mice, Knockout ,Heart ,Hypoxia-Inducible Factor 1, alpha Subunit ,Cell biology ,DNA-Binding Proteins ,Oxygen ,Tamoxifen ,Phenotype ,Endocrinology ,medicine.symptom ,Cardiology and Cardiovascular Medicine - Abstract
Background— Prolyl hydroxylase domain (PHD) proteins, including PHD1, PHD2, and PHD3, mediate oxygen-dependent degradation of hypoxia-inducible factor (HIF)-α subunits. Although angiogenic roles of hypoxia-inducible factors are well known, the roles of PHDs in the vascular system remain to be established. Methods and Results— We evaluated angiogenic phenotypes in mice carrying targeted disruptions in genes encoding different PHD isoforms. Although Phd1 −/− and Phd3 −/− mice did not display apparent angiogenic defects, broad-spectrum conditional knockout of Phd2 led to hyperactive angiogenesis and angiectasia. Blood vessels in PHD2-deficient mice were highly perfusable. Furthermore, examination of medium-sized vessels in subendocardial layer in the heart demonstrated successful recruitment of vascular smooth muscle cells. Surprisingly, increased vascular growth was independent of local efficiency of Phd2 disruption. Mice carrying significant Phd2 disruption in multiple organs, including the liver, heart, kidney, and lung, displayed excessive vascular growth not only in these organs but also in the brain, where Phd2 disruption was very inefficient. More surprisingly, increased accumulation of hypoxia-inducible factor-1α and angiectasia in the liver were not accompanied by corresponding increases in hepatic expression of Vegfa or angiopoietin-1 . However, the serum vascular endothelial growth factor-A level was significantly increased in PHD2-deficient mice. Conclusions— PHD2, but not PHD1 and PHD3, is a major negative regulator for vascular growth in adult mice. Increased angiogenesis in PHD2-deficient mice may be mediated by a novel systemic mechanism.
- Published
- 2007
- Full Text
- View/download PDF
43. Prolyl Hydroxylase Domain 2 Protein Suppresses Hypoxia-Induced Endothelial Cell Proliferation
- Author
-
Guo-Hua Fong and Kotaro Takeda
- Subjects
Vascular Endothelial Growth Factor A ,medicine.medical_specialty ,medicine.medical_treatment ,Procollagen-Proline Dioxygenase ,Biology ,Hydroxylation ,Transfection ,Cell Line ,Hypoxia-Inducible Factor-Proline Dioxygenases ,Immediate-Early Proteins ,chemistry.chemical_compound ,Downregulation and upregulation ,Internal medicine ,Internal Medicine ,medicine ,Gene silencing ,Gene Silencing ,Hypoxia ,Cell Proliferation ,Feedback, Physiological ,Cell growth ,Growth factor ,Endothelial Cells ,Hypoxia-Inducible Factor 1, alpha Subunit ,Up-Regulation ,Cell biology ,DNA-Binding Proteins ,Vascular endothelial growth factor ,Endothelial stem cell ,Endocrinology ,Hypoxia-inducible factors ,chemistry ,Mutation ,Procollagen-proline dioxygenase - Abstract
Prolyl hydroxylase domain 2 protein (PHD2) signals the degradation of hypoxia-inducible factor (HIF)-1α by hydroxylating specific prolyl residues located within oxygen-dependent degradation domains. As expected, endothelial cells (ECs) overexpressing PHD2 had reduced HIF-1α and vascular endothelial growth factor-A expression and failed to accelerate their proliferation in response to hypoxia. Surprisingly, although these cells displayed further reductions in HIF-1α and vascular endothelial growth factor-A expression when cultured under normoxia, there was no further reduction in EC proliferation. Thus, there seemed to be no consistent correlation between PHD2 hydroxylase–mediated suppression of HIF-1α expression and inhibition of EC growth. Indeed, overexpression of a mutant PHD2 lacking hydroxylase activity also greatly diminished EC response to hypoxia-induced increase in proliferation, in spite of the fact that hypoxia-induced HIF-1α accumulation was not affected by mutant PHD2. These data strongly suggest the existence of a hydroxylase-independent mechanism for PHD2-mediated inhibition of EC proliferation under hypoxia. In support of a physiological relevance of PHD2 overexpression, we found that endogenous PHD2 expression was significantly upregulated by hypoxia and that silencing of the Phd2 gene by RNA interference significantly enhanced hypoxia-induced EC proliferation. In conclusion, this study demonstrates that PHD2 may act as a negative feedback regulator to antagonize hypoxia-induced EC proliferation.
- Published
- 2007
- Full Text
- View/download PDF
44. Vasculogenesis and Angiogenesis in VEGF Receptor-1 Deficient Mice
- Author
-
Vivienne C. Ho and Guo-Hua Fong
- Subjects
Placental growth factor ,Angiogenesis ,Neovascularization, Physiologic ,Biology ,Endothelial cell differentiation ,Receptor tyrosine kinase ,Article ,chemistry.chemical_compound ,Mice ,Vasculogenesis ,Conditional gene knockout ,Animals ,Receptor ,Alleles ,Mice, Knockout ,Vascular Endothelial Growth Factor Receptor-1 ,Cell biology ,Vascular endothelial growth factor ,Germ Cells ,chemistry ,Animals, Newborn ,Genetic Loci ,Immunology ,Gene Targeting ,Models, Animal ,Mutation ,biology.protein ,cardiovascular system - Abstract
Vascular endothelial growth factor receptor-1 (VEGFR-1)/Flt-1 is a transmembrane tyrosine kinase receptor for VEGF-A, VEGF-B, and placental growth factor (PlGF). VEGFR-1 is an enigmatic molecule whose precise role in postnatal angiogenesis remains controversial. Although many postnatal and adult studies have been performed by manipulating VEGFR-1 ligands, including competitive binding by truncated VEGFR-1 protein, neutralization by antibodies, or specific ligand overexpression or knockout, much less is known at the level of the receptor per se, especially in vivo. Perplexingly, while VEGFR-1 negatively regulates endothelial cell differentiation during development, it has been implied in promoting angiogenesis under certain conditions in adult tissues, especially in tumors and ischemic tissues. Additionally, it is unclear how VEGFR-1 is involved in vascular maturation and maintenance of vascular quiescence in adult tissues. To facilitate further investigation, we generated a conditional knockout mouse line for VEGFR-1 and characterized angiogenesis in postnatal and adult mice, including angiogenesis in ischemic myocardium. These methods are briefly outlined in this chapter. We also discuss these findings in the context of the interplay between VEGF family members and their receptors, and summarize various mouse models in the VEGF pathway.
- Published
- 2015
45. Targeting of the pulmonary capillary vascular niche promotes lung alveolar repair and ameliorates fibrosis
- Author
-
Shahin Rafii, Michael Ginsberg, Sina Y. Rabbany, Thomas P. Sakmar, Deebly Chavez, Raphael Lis, Zhongwei Cao, Guo-Hua Fong, Bi-Sen Ding, and Koji Shido
- Subjects
0301 basic medicine ,Pathology ,Pulmonary Circulation ,Angiogenesis ,Pulmonary Fibrosis ,Fluorescent Antibody Technique ,chemistry.chemical_compound ,Mice ,Fibrosis ,Pulmonary fibrosis ,Serrate-Jagged Proteins ,RNA, Small Interfering ,Lung ,Wnt Signaling Pathway ,Antibiotics, Antineoplastic ,Receptors, Notch ,General Medicine ,Lung Injury ,respiratory system ,medicine.anatomical_structure ,Intercellular Signaling Peptides and Proteins ,Oligopeptides ,medicine.medical_specialty ,Notch signaling pathway ,Lung injury ,Biology ,Pulmonary Artery ,Bleomycin ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,medicine ,Animals ,Humans ,Regeneration ,Smad3 Protein ,Receptors, CXCR ,Vascular Endothelial Growth Factor Receptor-1 ,Regeneration (biology) ,Macrophages ,Calcium-Binding Proteins ,Endothelial Cells ,Membrane Proteins ,Fibroblasts ,medicine.disease ,Capillaries ,030104 developmental biology ,chemistry ,Hydrochloric Acid ,Jagged-1 Protein - Abstract
Although the lung can undergo self-repair after injury, fibrosis in chronically injured or diseased lungs can occur at the expense of regeneration. Here we study how a hematopoietic-vascular niche regulates alveolar repair and lung fibrosis. Using intratracheal injection of bleomycin or hydrochloric acid in mice, we show that repetitive lung injury activates pulmonary capillary endothelial cells (PCECs) and perivascular macrophages, impeding alveolar repair and promoting fibrosis. Whereas the chemokine receptor CXCR7, expressed on PCECs, acts to prevent epithelial damage and ameliorate fibrosis after a single round of treatment with bleomycin or hydrochloric acid, repeated injury leads to suppression of CXCR7 expression and recruitment of vascular endothelial growth factor receptor 1 (VEGFR1)-expressing perivascular macrophages. This recruitment stimulates Wnt/β-catenin-dependent persistent upregulation of the Notch ligand Jagged1 (encoded by Jag1) in PCECs, which in turn stimulates exuberant Notch signaling in perivascular fibroblasts and enhances fibrosis. Administration of a CXCR7 agonist or PCEC-targeted Jag1 shRNA after lung injury promotes alveolar repair and reduces fibrosis. Thus, targeting of a maladapted hematopoietic-vascular niche, in which macrophages, PCECs and perivascular fibroblasts interact, may help to develop therapy to spur lung regeneration and alleviate fibrosis.
- Published
- 2015
46. Potential Contributions of Intimal and Plaque Hypoxia to Atherosclerosis
- Author
-
Guo-Hua Fong
- Subjects
Endothelium ,Angiogenesis ,Inflammation ,Muscle, Smooth, Vascular ,medicine ,Animals ,Humans ,Foam cell ,business.industry ,Macrophages ,Monocyte ,Hypoxia (medical) ,Atherosclerosis ,medicine.disease ,Cell biology ,Lipoproteins, LDL ,medicine.anatomical_structure ,Vasa vasorum ,Immunology ,lipids (amino acids, peptides, and proteins) ,Endothelium, Vascular ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,business ,Infiltration (medical) ,Foam Cells - Abstract
Injury of arterial endothelium by abnormal shear stress and other insults induces migration and proliferation of vascular smooth muscle cells (VSMCs), which in turn leads to intimal thickening, hypoxia, and vasa vasorum angiogenesis. The resultant new blood vessels extend from the tunica media into the outer intima, allowing blood-borne oxidized low-density lipoprotein (oxLDL) particles to accumulate in outer intimal tissues by extravasation through local capillaries. In response to oxLDL accumulation, monocytes infiltrate into arterial wall tissues, where they differentiate into macrophages and subsequently evolve into foam cells by uptaking large quantities of oxLDL particles, the latter process being stimulated by hypoxia. Increased oxygen demand due to expanding macrophage and foam cell populations contributes to persistent hypoxia in plaque lesions, whereas hypoxia further promotes plaque growth by stimulating angiogenesis, monocyte infiltration, and oxLDL uptake into macrophages. Molecularly, the accumulation of hypoxia-inducible factor (HIF)-1α and the expression of its target genes mediate many of the hypoxia-induced processes during plaque initiation and growth. It is hoped that further understanding of the underlying mechanisms may lead to novel therapies for effective intervention of atherosclerosis.
- Published
- 2015
- Full Text
- View/download PDF
47. Hypoxia Signaling Cascade for Erythropoietin Production in Hepatocytes
- Author
-
Xiaoqing Pan, Hiroki Sekine, Yutaka Tojo, Norihiko Takeda, Ikuo Hirano, Tomokazu Souma, Kotaro Takeda, Masakazu Ichinose, Shinichi Kawaguchi, Takashi Dan, Guo-Hua Fong, Tadayuki Tsujita, Norio Suzuki, Toshio Miyata, and Masayuki Yamamoto
- Subjects
Gene isoform ,Transcription, Genetic ,Procollagen-Proline Dioxygenase ,Polycythemia ,Hypoxia-Inducible Factor-Proline Dioxygenases ,Mice ,hemic and lymphatic diseases ,Cell Line, Tumor ,Oxygen homeostasis ,medicine ,Basic Helix-Loop-Helix Transcription Factors ,Animals ,Humans ,Enhancer ,Molecular Biology ,Transcription factor ,Erythropoietin ,Mice, Knockout ,Kidney ,biology ,Anemia ,Cell Biology ,Articles ,Hypoxia-Inducible Factor 1, alpha Subunit ,Cell Hypoxia ,Chromatin ,Cell biology ,Enzyme Activation ,Fatty Liver ,Repressor Proteins ,Histone ,medicine.anatomical_structure ,Biochemistry ,Liver ,biology.protein ,Hepatocytes ,Apoptosis Regulatory Proteins ,medicine.drug ,Signal Transduction ,Transcription Factors - Abstract
Erythropoietin (Epo) is produced in the kidney and liver in a hypoxia-inducible manner via the activation of hypoxia-inducible transcription factors (HIFs) to maintain oxygen homeostasis. Accelerating Epo production in hepatocytes is one plausible therapeutic strategy for treating anemia caused by kidney diseases. To elucidate the regulatory mechanisms of hepatic Epo production, we analyzed mouse lines harboring liver-specific deletions of genes encoding HIF-prolyl-hydroxylase isoforms (PHD1, PHD2, and PHD3) that mediate the inactivation of HIF1α and HIF2α under normal oxygen conditions. The loss of all PHD isoforms results in both polycythemia, which is caused by Epo overproduction, and fatty livers. We found that deleting any combination of two PHD isoforms induces polycythemia without steatosis complications, whereas the deletion of a single isoform induces no apparent phenotype. Polycythemia is prevented by the loss of either HIF2α or the hepatocyte-specific Epo gene enhancer (EpoHE). Chromatin analyses show that the histones around EpoHE dissociate from the nucleosome structure after HIF2α activation. HIF2α also induces the expression of HIF3α, which is involved in the attenuation of Epo production. These results demonstrate that the total amount of PHD activity is more important than the specific function of each isoform for hepatic Epo expression regulated by a PHD-HIF2α-EpoHE cascade in vivo.
- Published
- 2015
48. Deficiency in the p110α subunit of PI3K results in diminished Tie2 expression and Tie2-/-–like vascular defects in mice
- Author
-
Robert L. Nussbaum, Li-Juan Duan, Etienne Lelievre, Pierre-Marie Bourbon, and Guo-Hua Fong
- Subjects
Endothelium ,Immunology ,Biology ,P110α ,Hemostasis, Thrombosis, and Vascular Biology ,Biochemistry ,Mice ,Phosphatidylinositol 3-Kinases ,chemistry.chemical_compound ,medicine ,Animals ,Cells, Cultured ,Mice, Knockout ,Endothelial Cells ,Gene Expression Regulation, Developmental ,Cell Biology ,Hematology ,Embryo, Mammalian ,Tunica intima ,Receptor, TIE-2 ,Cell biology ,Vascular endothelial growth factor B ,Vascular endothelial growth factor ,Protein Subunits ,Vascular endothelial growth factor A ,medicine.anatomical_structure ,Vascular endothelial growth factor C ,chemistry ,embryonic structures ,cardiovascular system ,Signal transduction - Abstract
Phosphoinositide 3-kinase (PI3K) is activated by transmembrane tyrosine kinases such as vascular endothelial growth factor (VEGF) receptors and Tie2 (tunica intima endothelial kinase 2), both of which are key regulators of vascular development. However, the in vivo role of PI3K during developmental vascularization remains to be defined. Here we demonstrate that mice deficient in the p110α catalytic subunit of PI3K display multiple vascular defects, including dilated vessels in the head, reduced branching morphogenesis in the endocardium, lack of hierarchical order of large and small branches in the yolk sac, and impaired development of anterior cardinal veins. These vascular defects are strikingly similar to those in mice defective in the Tie2 signaling pathway. Indeed, Tie2 protein levels were significantly lower in p110α-deficient mice. Furthermore, RNA interference of p110α in cultured endothelial cells significantly reduced Tie2 protein levels. These findings raise the possibility that PI3K may function as an upstream regulator of Tie2 expression during mouse development.
- Published
- 2005
- Full Text
- View/download PDF
49. Knockin of SV40 Tag oncogene in a mouse adenocarcinoma of the prostate model demonstrates advantageous features over the transgenic model
- Author
-
Manal Y. Gabril, Wenming Duan, Guo-Hua Fong, Jim W. Xuan, Franky L. Chan, Madeleine Moussa, and Hideki Sakai
- Subjects
Male ,Genetically modified mouse ,PCA3 ,Cancer Research ,Antigens, Polyomavirus Transforming ,Mice, Transgenic ,Simian virus 40 ,Adenocarcinoma ,Biology ,medicine.disease_cause ,Basement Membrane ,Transgenic Model ,Mice ,Prostate cancer ,Prostate ,Genetics ,medicine ,Animals ,Neoplasm Metastasis ,Molecular Biology ,Prostatic Neoplasms ,Prostatic Secretory Proteins ,Cancer ,Exons ,medicine.disease ,medicine.anatomical_structure ,Immunology ,Cancer research ,Carcinogenesis ,Orchiectomy - Abstract
Prostate cancer (CaP) is the most common cancer in adult men in North America. Since there is no naturally occurring prostate cancer in the mouse, preclinical studies stipulate for the establishment of a genetically manipulated mouse CaP model with features close to the human situation. In view of the limitations of transgenic technique-derived CaP models, herein we report the first application of knockin technology to establish a new mouse adenocarcinoma prostate model (PSP-KIMAP) by targeting of SV40 Tag to a prostate tissue-specific gene, PSP94 (prostate secretory protein of 94 amino acids). In order to demonstrate its novelty, we compared KIMAP to a PSP94 gene-directed transgenic mouse adenocarcinoma of the prostate (PSP-TGMAP) model. The CaP development of the PSP-KIMAP mice started almost immediately after puberty at 10 weeks of age from mouse prostatic intraepithelial neoplasia (mPIN) with microinvasion to well-differentiated CaP, and demonstrated a close-to-human kinetics of prolonged tumor growth and a predominance of well and moderately differentiated tumors. The invasive nature of KIMAP model was demonstrated by multitissue metastases (lymph node, lung and liver etc) and also by immunohistochemical study of multiple invasive prostate tumor markers. PSP-KIMAP model is responsive to androgen deprivation (castration). The knockin technology in our KIMAP model demonstrates highly predictive CaP development procedures and many advantageous features, which the traditional transgenic technique-derived CaP models could not reach for both basic and clinical studies. These features include the high stability of both phenotype and genotype, highly synchronous prostate cancer development, high and precise prostate tissue targeting and with no founder line variation. The differences between the two CaP models were attributed to the introduction of a single endogenous knockin mutation, resulting in a CaP model self-regulated and controlled by a prostate gene promoter/enhancer of PSP94.
- Published
- 2005
- Full Text
- View/download PDF
50. A hierarchical order of factors in the generation of FLK1- and SCL-expressing hematopoietic and endothelial progenitors from embryonic stem cells
- Author
-
Elizabeth Arentson, Wen Jie Zhang, Iva Afrikanova, Alexander Rosendahl, Guo-Hua Fong, Yun Shin Chung, Kyunghee Choi, and Changwon Park
- Subjects
Vascular Endothelial Growth Factor A ,Smad6 Protein ,Mitogen-Activated Protein Kinase 3 ,MAP Kinase Kinase 1 ,Smad Proteins ,Bone Morphogenetic Protein 4 ,Culture Media, Serum-Free ,Receptor tyrosine kinase ,Mice ,chemistry.chemical_compound ,Transforming Growth Factor beta ,Basic Helix-Loop-Helix Transcription Factors ,Enzyme Inhibitors ,Cells, Cultured ,T-Cell Acute Lymphocytic Leukemia Protein 1 ,Mitogen-Activated Protein Kinase 1 ,Extracellular Matrix Proteins ,Nonmuscle Myosin Type IIB ,biology ,Stem Cells ,Cell biology ,DNA-Binding Proteins ,Platelet Endothelial Cell Adhesion Molecule-1 ,Vascular endothelial growth factor ,Endothelial stem cell ,Haematopoiesis ,Bone morphogenetic protein 4 ,Bone Morphogenetic Proteins ,CD4 Antigens ,embryonic structures ,Mitogen-Activated Protein Kinases ,Smad5 Protein ,animal structures ,Smad1 Protein ,Transforming Growth Factor beta1 ,Proto-Oncogene Proteins ,Nitriles ,Butadienes ,Animals ,Humans ,Progenitor cell ,Molecular Biology ,Mitogen-Activated Protein Kinase Kinases ,Myosin Heavy Chains ,Hematopoietic Stem Cells ,Phosphoproteins ,Vascular Endothelial Growth Factor Receptor-2 ,Embryonic stem cell ,chemistry ,Trans-Activators ,biology.protein ,Endothelium, Vascular ,Transcription Factors ,Developmental Biology - Abstract
The receptor tyrosine kinase FLK1 and the transcription factor SCL play crucial roles in the establishment of hematopoietic and endothelial cell lineages in mice. We have previously used an in vitro differentiation model of embryonic stem (ES) cells and demonstrated that hematopoietic and endothelial cells develop via sequentially generated FLK1(+) and SCL(+) cells. To gain a better understanding of cellular and molecular events leading to hematopoietic specification, we examined factors necessary for FLK1(+) and SCL(+) cell induction in serum-free conditions. We demonstrate that bone morphogenetic protein (BMP) 4 was required for the generation of FLK1(+) and SCL(+) cells, and that vascular endothelial growth factor (VEGF) was necessary for the expansion and differentiation of SCL-expressing hematopoietic progenitors. Consistently, Flk1-deficient ES cells responded to BMP4 and generated TER119(+) and CD31(+) cells, but they failed to expand in response to VEGF. The Smad1/5 and map kinase pathways were activated by BMP4 and VEGF, respectively. The overexpression of SMAD6 in ES cells resulted in a reduction of FLK1(+) cells. In addition, a MAP kinase kinase 1 specific inhibitor blocked the expansion of SCL(+) cells in response to VEGF. Finally, VEGF mediated expansion of hematopoietic and endothelial cell progenitors was inhibited by TGFbeta1, but was augmented by activin A. Our studies suggest that hematopoietic and endothelial commitment from the mesoderm occurs via BMP4-mediated signals and that expansion and/or differentiation of such progenitors is achieved by an interplay of VEGF, TGFbeta1 and activin A signaling.
- Published
- 2004
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.