Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 190
Filter
1.
Nat Rev Mol Cell Biol ; 24(11): 816-834, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37491579

ABSTRACT

The formation of new blood vessels, called angiogenesis, is an essential pathophysiological process in which several families of regulators have been implicated. Among these, vascular endothelial growth factor A (VEGFA; also known as VEGF) and its two tyrosine kinase receptors, VEGFR1 and VEGFR2, represent a key signalling pathway mediating physiological angiogenesis and are also major therapeutic targets. VEGFA is a member of the gene family that includes VEGFB, VEGFC, VEGFD and placental growth factor (PLGF). Three decades after its initial isolation and cloning, VEGFA is arguably the most extensively investigated signalling system in angiogenesis. Although many mediators of angiogenesis have been identified, including members of the FGF family, angiopoietins, TGFß and sphingosine 1-phosphate, all current FDA-approved anti-angiogenic drugs target the VEGF pathway. Anti-VEGF agents are widely used in oncology and, in combination with chemotherapy or immunotherapy, are now the standard of care in multiple malignancies. Anti-VEGF drugs have also revolutionized the treatment of neovascular eye disorders such as age-related macular degeneration and ischaemic retinal disorders. In this Review, we emphasize the molecular, structural and cellular basis of VEGFA action as well as recent findings illustrating unexpected interactions with other pathways and provocative reports on the role of VEGFA in regenerative medicine. We also discuss clinical and translational aspects of VEGFA. Given the crucial role that VEGFA plays in regulating angiogenesis in health and disease, this molecule is largely the focus of this Review.


Subject(s)
Neoplasms , Vascular Endothelial Growth Factor A , Female , Humans , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Placenta Growth Factor , Angiogenesis Inhibitors/therapeutic use , Neoplasms/drug therapy , Neoplasms/genetics , Biology
2.
Cell ; 176(6): 1248-1264, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30849371

ABSTRACT

The discovery of vascular endothelial-derived growth factor (VEGF) has revolutionized our understanding of vasculogenesis and angiogenesis during development and physiological homeostasis. Over a short span of two decades, our understanding of the molecular mechanisms by which VEGF coordinates neurovascular homeostasis has become more sophisticated. The central role of VEGF in the pathogenesis of diverse cancers and blinding eye diseases has also become evident. Elucidation of the molecular regulation of VEGF and the transformative development of multiple therapeutic pathways targeting VEGF directly or indirectly is a powerful case study of how fundamental research can guide innovation and translation. It is also an elegant example of how agnostic discovery and can transform our understanding of human disease. This review will highlight critical nodal points in VEGF biology, including recent developments in immunotherapy for cancer and multitarget approaches in neovascular eye disease.


Subject(s)
Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/physiology , Vascular Endothelial Growth Factors/physiology , Animals , Humans , Immunotherapy/methods , Neoplasms/drug therapy , Neoplasms/metabolism , Retinal Vessels/metabolism , Retinal Vessels/physiology , Signal Transduction/physiology , Vascular Endothelial Growth Factors/metabolism
3.
Cell ; 167(1): 275-284.e6, 2016 Sep 22.
Article in English | MEDLINE | ID: mdl-27662093

ABSTRACT

The VEGF-A isoforms play a crucial role in vascular development, and the VEGF signaling pathway is a clinically validated therapeutic target for several pathological conditions. Alternative mRNA splicing leads to the generation of multiple VEGF-A isoforms, including VEGF165. A recent study reported the presence of another isoform, VEGF-Ax, arising from programmed readthrough translation. Compared to VEGF165, VEGF-Ax has a 22-amino-acid extension in the COOH terminus and has been reported to function as a negative regulator of VEGF signaling in endothelial cells, with potent anti-angiogenic effects. Here, we show that, contrary to the earlier report, VEGF-Ax stimulates endothelial cell mitogenesis, angiogenesis, as well as vascular permeability. Accordingly, VEGF-Ax induces phosphorylation of key tyrosine residues in VEGFR-2. Notably, VEGF-Ax was less potent than VEGF165, consistent with its impaired binding to the VEGF co-receptor neuropilin-1.


Subject(s)
Neovascularization, Physiologic/physiology , Vascular Endothelial Growth Factor A , Alternative Splicing , Amino Acid Sequence , Angiogenesis Inducing Agents/pharmacology , Angiogenesis Inhibitors/pharmacology , Animals , Capillary Permeability/genetics , Capillary Permeability/physiology , Chemotaxis/drug effects , Cloning, Molecular , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/physiology , Guinea Pigs , HEK293 Cells , Humans , Mice , Mitogens/pharmacology , Mitosis/drug effects , Mitosis/physiology , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/genetics , Neuropilin-1/metabolism , Protein Biosynthesis , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Tyrosine/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
4.
Cell ; 165(4): 882-95, 2016 May 05.
Article in English | MEDLINE | ID: mdl-27133169

ABSTRACT

High-fat diet (HFD) feeding induces rapid reprogramming of systemic metabolism. Here, we demonstrate that HFD feeding of mice downregulates glucose transporter (GLUT)-1 expression in blood-brain barrier (BBB) vascular endothelial cells (BECs) and reduces brain glucose uptake. Upon prolonged HFD feeding, GLUT1 expression is restored, which is paralleled by increased expression of vascular endothelial growth factor (VEGF) in macrophages at the BBB. In turn, inducible reduction of GLUT1 expression specifically in BECs reduces brain glucose uptake and increases VEGF serum concentrations in lean mice. Conversely, myeloid-cell-specific deletion of VEGF in VEGF(Δmyel) mice impairs BBB-GLUT1 expression, brain glucose uptake, and memory formation in obese, but not in lean mice. Moreover, obese VEGF(Δmyel) mice exhibit exaggerated progression of cognitive decline and neuroinflammation on an Alzheimer's disease background. These experiments reveal that transient, HFD-elicited reduction of brain glucose uptake initiates a compensatory increase of VEGF production and assign obesity-associated macrophage activation a homeostatic role to restore cerebral glucose metabolism, preserve cognitive function, and limit neurodegeneration in obesity.


Subject(s)
Brain/metabolism , Diet, High-Fat , Glucose/metabolism , Obesity/physiopathology , Vascular Endothelial Growth Factor A/metabolism , Animals , Blood-Brain Barrier/metabolism , Cognition , Endothelial Cells/metabolism , Fatty Acids/metabolism , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Mice , Myeloid Cells/metabolism
5.
Cell ; 152(5): 1065-76, 2013 Feb 28.
Article in English | MEDLINE | ID: mdl-23452854

ABSTRACT

Medulloblastoma is the most common pediatric malignant brain tumor. Although current therapies improve survival, these regimens are highly toxic and are associated with significant morbidity. Here, we report that placental growth factor (PlGF) is expressed in the majority of medulloblastomas, independent of their subtype. Moreover, high expression of PlGF receptor neuropilin 1 (Nrp1) correlates with poor overall survival in patients. We demonstrate that PlGF and Nrp1 are required for the growth and spread of medulloblastoma: PlGF/Nrp1 blockade results in direct antitumor effects in vivo, resulting in medulloblastoma regression, decreased metastasis, and increased mouse survival. We reveal that PlGF is produced in the cerebellar stroma via tumor-derived Sonic hedgehog (Shh) and show that PlGF acts through Nrp1-and not vascular endothelial growth factor receptor 1-to promote tumor cell survival. This critical tumor-stroma interaction-mediated by Shh, PlGF, and Nrp1 across medulloblastoma subtypes-supports the development of therapies targeting PlGF/Nrp1 pathway.


Subject(s)
Cerebellar Neoplasms/pathology , Cerebellum/metabolism , Medulloblastoma/pathology , Neuropilin-1/metabolism , Pregnancy Proteins/metabolism , Signal Transduction , Animals , Cells, Cultured , Cerebellar Neoplasms/metabolism , Humans , Medulloblastoma/metabolism , Mice , Mice, Knockout , Neoplasm Transplantation , Paracrine Communication , Placenta Growth Factor , Transplantation, Heterologous , Vascular Endothelial Growth Factor Receptor-1/metabolism
6.
Cell ; 151(2): 384-99, 2012 Oct 12.
Article in English | MEDLINE | ID: mdl-23063127

ABSTRACT

Vascular endothelial growth factor and its receptors, FLK1/KDR and FLT1, are key regulators of angiogenesis. Unlike FLK1/KDR, the role of FLT1 has remained elusive. FLT1 is produced as soluble (sFLT1) and full-length isoforms. Here, we show that pericytes from multiple tissues produce sFLT1. To define the biologic role of sFLT1, we chose the glomerular microvasculature as a model system. Deletion of Flt1 from specialized glomerular pericytes, known as podocytes, causes reorganization of their cytoskeleton with massive proteinuria and kidney failure, characteristic features of nephrotic syndrome in humans. The kinase-deficient allele of Flt1 rescues this phenotype, demonstrating dispensability of the full-length isoform. Using cell imaging, proteomics, and lipidomics, we show that sFLT1 binds to the glycosphingolipid GM3 in lipid rafts on the surface of podocytes, promoting adhesion and rapid actin reorganization. sFLT1 also regulates pericyte function in vessels outside of the kidney. Our findings demonstrate an autocrine function for sFLT1 to control pericyte behavior.


Subject(s)
Kidney Glomerulus/cytology , Kidney Glomerulus/metabolism , Podocytes/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Animals , Gangliosides/metabolism , Humans , In Vitro Techniques , Lipid Metabolism , Lipids/chemistry , Mice , Mice, Transgenic , Pericytes/metabolism , Proteinuria/metabolism , Signal Transduction , Syndecans/metabolism , Vascular Endothelial Growth Factor Receptor-1/genetics
8.
Pharmacol Rev ; 75(6): 1167-1199, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37684054

ABSTRACT

The prokineticins (PKs) were discovered approximately 20 years ago as small peptides inducing gut contractility. Today, they are established as angiogenic, anorectic, and proinflammatory cytokines, chemokines, hormones, and neuropeptides involved in variety of physiologic and pathophysiological pathways. Their altered expression or mutations implicated in several diseases make them a potential biomarker. Their G-protein coupled receptors, PKR1 and PKR2, have divergent roles that can be therapeutic target for treatment of cardiovascular, metabolic, and neural diseases as well as pain and cancer. This article reviews and summarizes our current knowledge of PK family functions from development of heart and brain to regulation of homeostasis in health and diseases. Finally, the review summarizes the established roles of the endogenous peptides, synthetic peptides and the selective ligands of PKR1 and PKR2, and nonpeptide orthostatic and allosteric modulator of the receptors in preclinical disease models. The present review emphasizes the ambiguous aspects and gaps in our knowledge of functions of PKR ligands and elucidates future perspectives for PK research. SIGNIFICANCE STATEMENT: This review provides an in-depth view of the prokineticin family and PK receptors that can be active without their endogenous ligand and exhibits "constitutive" activity in diseases. Their non- peptide ligands display promising effects in several preclinical disease models. PKs can be the diagnostic biomarker of several diseases. A thorough understanding of the role of prokineticin family and their receptor types in health and diseases is critical to develop novel therapeutic strategies with safety concerns.


Subject(s)
Neoplasms , Neuropeptides , Humans , Receptors, G-Protein-Coupled/metabolism , Neuropeptides/metabolism , Peptides , Neoplasms/drug therapy , Biomarkers
9.
Physiol Rev ; 98(2): 1055-1082, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29537336

ABSTRACT

The mammalian prokineticins family comprises two conserved proteins, EG-VEGF/PROK1 and Bv8/PROK2, and their two highly related G protein-coupled receptors, PKR1 and PKR2. This signaling system has been linked to several important biological functions, including gastrointestinal tract motility, regulation of circadian rhythms, neurogenesis, angiogenesis and cancer progression, hematopoiesis, and nociception. Mutations in PKR2 or Bv8/PROK2 have been associated with Kallmann syndrome, a developmental disorder characterized by defective olfactory bulb neurogenesis, impaired development of gonadotropin-releasing hormone neurons, and infertility. Also, Bv8/PROK2 is strongly upregulated in neutrophils and other inflammatory cells in response to granulocyte-colony stimulating factor or other myeloid growth factors and functions as a pronociceptive mediator in inflamed tissues as well as a regulator of myeloid cell-dependent tumor angiogenesis. Bv8/PROK2 has been also implicated in neuropathic pain. Anti-Bv8/PROK2 antibodies or small molecule PKR inhibitors ameliorate pain arising from tissue injury and inhibit angiogenesis and inflammation associated with tumors or some autoimmune disorders.


Subject(s)
Inflammation Mediators/metabolism , Inflammation/metabolism , Myeloid Cells/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Movement/physiology , Humans , Neurotransmitter Agents/metabolism
10.
Cell ; 140(2): 268-79, 2010 Jan 22.
Article in English | MEDLINE | ID: mdl-20141840

ABSTRACT

It is established that tumor cell-derived VEGF acts on endothelial cells to promote angiogenesis and tumor growth. Here, we demonstrate that in K5-SOS-dependent mouse skin tumors, autocrine VEGF is required for tumor cell proliferation in a cell-autonomous and angiogenesis-independent manner. VEGF is upregulated in SOS-expressing tumors, and its deletion in epidermal cells delays tumorigenesis by suppressing angiogenesis and tumor cell proliferation. Epidermis-specific Flt1 deletion also impairs tumorigenesis and proliferation. Surprisingly, complete tumor inhibition occurs in the absence of VEGF in EGFR mutant mice, demonstrating that VEGFR and EGFR synergize in neoplastic cells to promote tumor growth. Mechanistically, K5-SOS upregulates VEGF, Flt1, and Neuropilin-1 in an Erk-dependent manner, thereby activating an autocrine proliferation loop, whereas EGFR prevents tumor cells from apoptosis. Moreover, Flt1 is upregulated in human SCC, and its inhibition in SCC cells impairs proliferation. Thus, in addition to regulating angiogenesis, VEGF has to be considered as a potent growth factor for epidermal tumors.


Subject(s)
Carcinoma, Squamous Cell/metabolism , ErbB Receptors/metabolism , Skin Neoplasms/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Autocrine Communication , Cell Line, Tumor , Cells, Cultured , ErbB Receptors/genetics , Humans , Mice , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism
11.
Cell ; 141(1): 166-77, 2010 Apr 02.
Article in English | MEDLINE | ID: mdl-20371352

ABSTRACT

It has been recently reported that treatment with an anti-placenta growth factor (PlGF) antibody inhibits metastasis and primary tumor growth. Here we show that, although anti-PlGF treatment inhibited wound healing, extravasation of B16F10 cells, and growth of a tumor engineered to overexpress the PlGF receptor (VEGFR-1), neutralization of PlGF using four novel blocking antibodies had no significant effect on tumor angiogenesis in 15 models. Also, genetic ablation of the tyrosine kinase domain of VEGFR-1 in the host did not result in growth inhibition of the anti-VEGF-A sensitive or resistant tumors tested. Furthermore, combination of anti-PlGF with anti-VEGF-A antibodies did not result in greater antitumor efficacy than anti-VEGF-A monotherapy. In conclusion, our data argue against an important role of PlGF during primary tumor growth in most models and suggest that clinical evaluation of anti-PlGF antibodies may be challenging.


Subject(s)
Neoplasms/blood supply , Neovascularization, Pathologic , Pregnancy Proteins/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Cell Line, Tumor , Humans , Mice , Mice, Inbred BALB C , Placenta Growth Factor , Pregnancy Proteins/antagonists & inhibitors , Vascular Endothelial Growth Factors
12.
Annu Rev Cell Dev Biol ; 27: 563-84, 2011.
Article in English | MEDLINE | ID: mdl-21756109

ABSTRACT

The formation of the vascular network is an intricate and complex process that is an obligate requirement during vertebrate development. The cardiovascular system is the first organ to develop and reach a functional state, which underscores the crucial role of the vasculature in the developing embryo. The development of the vasculature into highly branched conduits needs to occur in numerous sites and in precise patterns to supply oxygen and nutrients to the rapidly expanding tissue of the embryo. This process is mediated by the coordinated response of vascular endothelial and mural cells to the heterogeneous angiogenic cues provided by tissues and organs, whereas aberrant regulation and coordination of angiogenic signals during development result in lethality, impaired organ development, or disease states. This article reviews the essential signaling pathways required for establishment of the vertebrate vasculature with a major focus on a key regulatory factor, vascular endothelial growth factor (VEGF). We also discuss current knowledge of physiological angiogenic processes as well as their disruptions in pathological processes, particularly tumorigenesis.


Subject(s)
Cardiovascular System , Neovascularization, Pathologic/physiopathology , Neovascularization, Physiologic/physiology , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/metabolism , Animals , Bone and Bones/blood supply , Bone and Bones/physiology , Cardiovascular System/anatomy & histology , Cardiovascular System/growth & development , Cardiovascular System/metabolism , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Protein Isoforms/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Reproduction , Wound Healing
13.
Proc Natl Acad Sci U S A ; 118(21)2021 05 25.
Article in English | MEDLINE | ID: mdl-34006633

ABSTRACT

Neovascularization is a key feature of ischemic retinal diseases and the wet form of age-related macular degeneration (AMD), all leading causes of severe vision loss. Vascular endothelial growth factor (VEGF) inhibitors have transformed the treatment of these disorders. Millions of patients have been treated with these drugs worldwide. However, in real-life clinical settings, many patients do not experience the same degree of benefit observed in clinical trials, in part because they receive fewer anti-VEGF injections. Therefore, there is an urgent need to discover and identify novel long-acting VEGF inhibitors. We hypothesized that binding to heparan-sulfate proteoglycans (HSPG) in the vitreous, and possibly other ocular structures, may be a strategy to promote intraocular retention, ultimately leading to a reduced burden of intravitreal injections. We designed a series of VEGF receptor 1 variants and identified some with strong heparin-binding characteristics and ability to bind to vitreous matrix. Our data indicate that some of our variants have longer duration and greater efficacy in animal models of intraocular neovascularization than current standard of care. Our study represents a systematic attempt to exploit the functional diversity associated with heparin affinity of a VEGF receptor.


Subject(s)
Choroidal Neovascularization/drug therapy , Heparan Sulfate Proteoglycans/pharmacology , Macular Degeneration/drug therapy , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/genetics , Angiogenesis Inhibitors/chemistry , Animals , Cell Proliferation/drug effects , Choroidal Neovascularization/genetics , Choroidal Neovascularization/pathology , Crystallography, X-Ray , Endothelial Cells/drug effects , Eye/drug effects , Eye/pathology , Heparan Sulfate Proteoglycans/genetics , Heparan Sulfate Proteoglycans/immunology , Heparin/genetics , Human Umbilical Vein Endothelial Cells , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/pharmacology , Immunoglobulin Fc Fragments/ultrastructure , Intravitreal Injections , Macular Degeneration/genetics , Macular Degeneration/pathology , Mice , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/ultrastructure , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vitreous Body/drug effects
14.
Proc Natl Acad Sci U S A ; 117(35): 21598-21608, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32817421

ABSTRACT

We tested cis-ApcΔ716/Smad4+/- and cis-ApcΔ716/Smad4+/-KrasG12D mice, which recapitulate key genetic abnormalities accumulating during colorectal cancer (CRC) tumorigenesis in humans, for responsiveness to anti-VEGF therapy. We found that even tumors in cis-ApcΔ716/Smad4+/-KrasG12D mice, although highly aggressive, were suppressed by anti-VEGF treatment. We tested the hypothesis that inflammation, a major risk factor and trigger for CRC, may affect responsiveness to anti-VEGF. Chemically induced colitis (CIC) in cis-ApcΔ716/Smad4+/- and cis-ApcΔ716/Smad4+/-KrasG12D mice promoted development of colon tumors that were largely resistant to anti-VEGF treatment. The myeloid growth factor G-CSF was markedly increased in the serum after induction of colitis. Antibodies blocking G-CSF, or its target Bv8/PROK2, suppressed tumor progression and myeloid cell infiltration when combined with anti-VEGF in CIC-associated CRC and in anti-VEGF-resistant CRC liver metastasis models. In a series of CRC specimens, tumor-infiltrating neutrophils strongly expressed Bv8/PROK2. CRC patients had significantly higher plasma Bv8/PROK2 levels than healthy volunteers and high plasma Bv8/PROK2 levels were inversely correlated with overall survival. Our findings establish Bv8/PROK2 as a translational target in CRC, in combination with anti-VEGF agents.


Subject(s)
Colorectal Neoplasms/genetics , Neutrophils/drug effects , Neutrophils/metabolism , Angiogenesis Inducing Agents/metabolism , Animals , Antibodies/immunology , Colonic Neoplasms/metabolism , Colorectal Neoplasms/metabolism , Female , Granulocyte Colony-Stimulating Factor/metabolism , Liver Neoplasms/metabolism , Male , Mice , Mice, Inbred C57BL , Models, Genetic , Myeloid Cells/metabolism , Neovascularization, Pathologic/pathology , Vascular Endothelial Growth Factor A/drug effects , Vascular Endothelial Growth Factor A/immunology , Vascular Endothelial Growth Factor A/metabolism
15.
J Transl Med ; 20(1): 527, 2022 11 12.
Article in English | MEDLINE | ID: mdl-36371231

ABSTRACT

BACKGROUND: The role of the inflammatory milieu in prostate cancer progression is not well understood. Differences in inflammatory signaling between localized and metastatic disease may point to opportunities for early intervention. METHODS: We modeled PCa disease progression by analyzing RNA-seq of localized vs. metastatic patient samples, followed by CIBERSORTx to assess their immune cell populations. The VHA CDW registry of PCa patients was analyzed for anti-TNF clinical outcomes. RESULTS: We observed statistically significant opposing patterns of IL-6 and TNFα expression between localized and metastatic disease. IL-6 was robustly expressed in localized disease and downregulated in metastatic disease. The reverse was observed with TNFα expression. Metastatic disease was also characterized by downregulation of adhesion molecule E-selectin, matrix metalloproteinase ADAMTS-4 and a shift to M2 macrophages whereas localized disease demonstrated a preponderance of M1 macrophages. Treatment with anti-TNF agents was associated with earlier stage disease at diagnosis. CONCLUSIONS: Our data points to clearly different inflammatory contexts between localized and metastatic prostate cancer. Primary localized disease demonstrates local inflammation and adaptive immunity, whereas metastases are characterized by immune cold microenvironments and a shift towards resolution of inflammation and tissue repair. Therapies that interfere with these inflammatory networks may offer opportunities for early intervention in monotherapy or in combination with immunotherapies and anti-angiogenic approaches.


Subject(s)
Immune Evasion , Prostatic Neoplasms , Male , Humans , Interleukin-6 , Tumor Necrosis Factor-alpha , Tumor Necrosis Factor Inhibitors , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Inflammation , Tumor Microenvironment
16.
Proc Natl Acad Sci U S A ; 115(43): 11060-11065, 2018 10 23.
Article in English | MEDLINE | ID: mdl-30301793

ABSTRACT

The tumor-promoting functions of neutrophils have been mainly attributed to induction of tumor angiogenesis or suppression of anticancer immunity. However, a direct impact of neutrophils on tumor cell growth and metastasis remains largely uncharacterized. Here, we coupled a proteomic approach with a functional screen to interrogate the secretome of tumor-associated neutrophils. Surprisingly, the iron-transporting protein transferrin was identified as the major mitogen for tumor cells secreted by neutrophils. Depletion of neutrophils inhibited lung metastasis and transferrin production in the metastatic microenvironment. Deletion of transferrin receptor suppressed growth of lung-colonizing tumor cells. Also, media conditioned by neutrophils isolated from metastatic breast cancer patients stimulated growth of human breast cancer cells, an effect that was largely abolished by transferrin immunodepletion. We identified GM-CSF, which is produced primarily by tumor cells, as a selective inducer of de novo transferrin synthesis in neutrophils through the Jak/Stat5ß pathway. GM-CSF neutralization or inhibition of Jak kinases curtailed neutrophil transferrin expression in vitro and in vivo as well as cancer metastasis. Thus, transferrin provides a mechanistic link between neutrophils and metastatic growth owing to the ability of tumor-infiltrating neutrophils to locally deliver this growth-promoting protein in response to GM-CSF stimulation. Our study identifies neutrophil-derived transferrin as a key regulator of metastatic tumor cell growth and a therapeutic target for antimetastatic treatment.


Subject(s)
Cell Proliferation/physiology , Neoplasm Metastasis/pathology , Neutrophils/metabolism , Neutrophils/pathology , Transferrin/metabolism , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Receptors, Transferrin/metabolism
17.
Angiogenesis ; 22(2): 311-323, 2019 05.
Article in English | MEDLINE | ID: mdl-30539314

ABSTRACT

TH17 cells play important yet complex roles in cancer development and progression. We previously reported that TH17 cells and IL-17 mediate resistance to anti-VEGF therapy by inducing recruitment of immunosuppressive and proangiogenic myeloid cells to the tumor microenvironment. Here, we demonstrate that IL-22, a key effector cytokine expressed by TH17 cells, directly acts on endothelial cells to promote tumor angiogenesis. IL-22 induces endothelial cell proliferation, survival, and chemotaxis in vitro and neovascularization in an ex vivo mouse choroid explant model. Blockade of IL-22, with a neutralizing antibody, significantly inhibits tumor growth associated with reduced microvascular density. No synergistic effect of IL-22 with VEGF was observed. These results identify IL-22 as a potential therapeutic target for blocking tumor angiogenesis.


Subject(s)
Interleukins/physiology , Neoplasms/blood supply , Neovascularization, Pathologic/genetics , Animals , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Female , Hep G2 Cells , Homeodomain Proteins/genetics , Human Umbilical Vein Endothelial Cells , Humans , Interleukins/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Neoplasms/genetics , Neoplasms/pathology , Neovascularization, Pathologic/pathology , Th17 Cells/physiology , Vascular Endothelial Growth Factor A/pharmacology , Interleukin-22
18.
Nature ; 494(7436): 243-6, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-23334418

ABSTRACT

Vascular patterning is critical for organ function. In the eye, there is simultaneous regression of embryonic hyaloid vasculature (important to clear the optical path) and formation of the retinal vasculature (important for the high metabolic demands of retinal neurons). These events occur postnatally in the mouse. Here we have identified a light-response pathway that regulates both processes. We show that when mice are mutated in the gene (Opn4) for the atypical opsin melanopsin, or are dark-reared from late gestation, the hyaloid vessels are persistent at 8 days post-partum and the retinal vasculature overgrows. We provide evidence that these vascular anomalies are explained by a light-response pathway that suppresses retinal neuron number, limits hypoxia and, as a consequence, holds local expression of vascular endothelial growth factor (VEGFA) in check. We also show that the light response for this pathway occurs in late gestation at about embryonic day 16 and requires the photopigment in the fetus and not the mother. Measurements show that visceral cavity photon flux is probably sufficient to activate melanopsin-expressing retinal ganglion cells in the mouse fetus. These data thus show that light--the stimulus for function of the mature eye--is also critical in preparing the eye for vision by regulating retinal neuron number and initiating a series of events that ultimately pattern the ocular blood vessels.


Subject(s)
Eye/blood supply , Eye/growth & development , Fetus/radiation effects , Light Signal Transduction/radiation effects , Light , Retinal Neurons/radiation effects , Rod Opsins/metabolism , Animals , Cell Count , Cell Hypoxia/radiation effects , Eye/metabolism , Eye/radiation effects , Female , Fetus/cytology , Fetus/embryology , Fetus/metabolism , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic , Neovascularization, Physiologic/radiation effects , Photons , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/radiation effects , Retinal Neurons/cytology , Retinal Neurons/metabolism , Rod Opsins/deficiency , Rod Opsins/genetics , Vascular Endothelial Growth Factor A/metabolism
19.
Angiogenesis ; 21(3): 425-532, 2018 08.
Article in English | MEDLINE | ID: mdl-29766399

ABSTRACT

The formation of new blood vessels, or angiogenesis, is a complex process that plays important roles in growth and development, tissue and organ regeneration, as well as numerous pathological conditions. Angiogenesis undergoes multiple discrete steps that can be individually evaluated and quantified by a large number of bioassays. These independent assessments hold advantages but also have limitations. This article describes in vivo, ex vivo, and in vitro bioassays that are available for the evaluation of angiogenesis and highlights critical aspects that are relevant for their execution and proper interpretation. As such, this collaborative work is the first edition of consensus guidelines on angiogenesis bioassays to serve for current and future reference.


Subject(s)
Biological Assay/methods , Neoplasms , Neovascularization, Pathologic , Animals , Biological Assay/instrumentation , Guidelines as Topic , Humans , Mice , Neoplasms/blood supply , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology
20.
Development ; 142(4): 672-80, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25617432

ABSTRACT

Blood vessels serve as key regulators of organogenesis by providing oxygen, nutrients and molecular signals. During limb development, programmed cell death (PCD) contributes to separation of the digits. Interestingly, prior to the onset of PCD, the autopod vasculature undergoes extensive patterning that results in high interdigital vascularity. Here, we show that in mice, the limb vasculature positively regulates interdigital PCD. In vivo, reduction in interdigital vessel number inhibited PCD, resulting in syndactyly, whereas an increment in vessel number and distribution resulted in elevation and expansion of PCD. Production of reactive oxygen species (ROS), toxic compounds that have been implicated in PCD, also depended on interdigital vascular patterning. Finally, ex vivo incubation of limbs in gradually decreasing oxygen levels led to a correlated reduction in both ROS production and interdigital PCD. The results support a role for oxygen in these processes and provide a mechanistic explanation for the counterintuitive positive role of the vasculature in PCD. In conclusion, we suggest a new role for vascular patterning during limb development in regulating interdigital PCD by ROS production. More broadly, we propose a double safety mechanism that restricts PCD to interdigital areas, as the genetic program of PCD provides the first layer and vascular patterning serves as the second.


Subject(s)
Cell Death/physiology , Reactive Oxygen Species/metabolism , Animals , Extremities/blood supply , Extremities/embryology , Female , Gene Expression Regulation, Developmental/physiology , Mice , Organ Culture Techniques , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL