Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Med Chem ; 61(21): 9691-9721, 2018 11 08.
Article in English | MEDLINE | ID: mdl-30289716

ABSTRACT

HIF-2α, a member of the HIF family of transcription factors, is a key oncogenic driver in cancers such as clear cell renal cell carcinoma (ccRCC). A signature feature of these cancers is the overaccumulation of HIF-2α protein, often by inactivation of the E3 ligase VHL (von Hippel-Lindau). Herein we disclose our structure based drug design (SBDD) approach that culminated in the identification of PT2385, the first HIF-2α antagonist to enter clinical trials. Highlights include the use of a putative n → π*Ar interaction to guide early analog design, the conformational restriction of an essential hydroxyl moiety, and the remarkable impact of fluorination near the hydroxyl group. Evaluation of select compounds from two structural classes in a sequence of PK/PD, efficacy, PK, and metabolite profiling identified 10i (PT2385, luciferase EC50 = 27 nM) as the clinical candidate. Finally, a retrospective crystallographic analysis describes the structural perturbations necessary for efficient antagonism.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Carcinoma, Renal Cell/pathology , Drug Design , Indans/chemistry , Indans/pharmacology , Kidney Neoplasms/pathology , Sulfones/chemistry , Sulfones/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Basic Helix-Loop-Helix Transcription Factors/chemistry , Cell Line, Tumor , Dogs , Indans/pharmacokinetics , Mice , Models, Molecular , Protein Conformation , Rats , Structure-Activity Relationship , Sulfones/pharmacokinetics , Tissue Distribution
2.
Cancer Res ; 76(18): 5491-500, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27635045

ABSTRACT

More than 90% of clear cell renal cell carcinomas (ccRCC) exhibit inactivation of the von Hippel-Lindau (pVHL) tumor suppressor, establishing it as the major underlying cause of this malignancy. pVHL inactivation results in stabilization of the hypoxia-inducible transcription factors, HIF1α and HIF2α, leading to expression of a genetic program essential for the initiation and progression of ccRCC. Herein, we describe the potent, selective, and orally active small-molecule inhibitor PT2385 as a specific antagonist of HIF2α that allosterically blocks its dimerization with the HIF1α/2α transcriptional dimerization partner ARNT/HIF1ß. PT2385 inhibited the expression of HIF2α-dependent genes, including VEGF-A, PAI-1, and cyclin D1 in ccRCC cell lines and tumor xenografts. Treatment of tumor-bearing mice with PT2385 caused dramatic tumor regressions, validating HIF2α as a pivotal oncogenic driver in ccRCC. Notably, unlike other anticancer agents that inhibit VEGF receptor signaling, PT2385 exhibited no adverse effect on cardiovascular performance. Thus, PT2385 represents a novel class of therapeutics for the treatment of RCC with potent preclincal efficacy as well as improved tolerability relative to current agents that target the VEGF pathway. Cancer Res; 76(18); 5491-500. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , Animals , Antineoplastic Agents/chemistry , Calorimetry , Cell Line, Tumor , Crystallography, X-Ray , Humans , Immunohistochemistry , Immunoprecipitation , Mice , Mice, SCID , Polymerase Chain Reaction , Xenograft Model Antitumor Assays
3.
J Neurotrauma ; 29(3): 579-88, 2012 Feb 10.
Article in English | MEDLINE | ID: mdl-21933012

ABSTRACT

Manipulation of various components of the endoplasmic reticulum (ER) stress response (ERSR) has led to functional recovery in diabetes, cancer, and several neurodegenerative diseases, indicating its use as a potential therapeutic intervention. One of the downstream pro-apoptotic transcription factors activated by the ERSR is CCAAT enhancer binding protein (C/EBP) homologous protein (CHOP). Recently, we showed significant recovery in hindlimb locomotion function after moderate contusive spinal cord injury (SCI) in mice null for CHOP. However, more than 40% of human SCI are complete. Thus the present study examined the potential therapeutic modulation of CHOP in a more severe SCI injury. Contused wild-type spinal cords showed a rapid activation of PERK, ATF6, and IRE-1, the three arms of the ERSR signaling pathway, specifically at the injury epicenter. Confocal images of phosphorylated EIF2α, GRP78, CHOP, ATF4, and GADD34 localized the activation of the ERSR in neurons and oligodendrocytes at the injury epicenter. To directly determine the role of CHOP, wild-type and CHOP-null mice with severe contusive SCI were analyzed for improvement in hindlimb locomotion. Despite the loss of CHOP, the other effectors in the ERSR pathway were significantly increased beyond that observed previously with moderate injury. Concomitantly, Basso Mouse Scale (BMS) scores and white matter sparing between the wild-type and CHOP-null mice revealed no significant differences. Given the complex pathophysiology of severe SCI, ablation of CHOP alone is not sufficient to rescue functional deficits. These data raise the caution that injury severity may be a key variable in attempting to translate preclinical therapies to clinical practice.


Subject(s)
Apoptosis/physiology , Endoplasmic Reticulum/physiology , Locomotion/physiology , Spinal Cord Injuries/physiopathology , Transcription Factor CHOP/physiology , Animals , Behavior, Animal/physiology , Blotting, Western , Claudins , Contusions/pathology , Contusions/physiopathology , Endoplasmic Reticulum Chaperone BiP , Female , Heat-Shock Proteins/metabolism , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin Basic Protein/metabolism , Nerve Tissue Proteins/metabolism , Protein Phosphatase 1/metabolism , RNA/biosynthesis , RNA/isolation & purification , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Spinal Cord/pathology , Spinal Cord Injuries/pathology , Transcription Factor CHOP/genetics
4.
Glia ; 59(10): 1489-502, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21638341

ABSTRACT

Activation of the unfolded protein response (UPR) is involved in the pathogenesis of numerous CNS myelin abnormalities; yet, its direct role in traumatic spinal cord injury (SCI)-induced demyelination is not known. The UPR is an evolutionarily conserved cell defense mechanism initiated to restore endoplasmic reticulum homeostasis in response to various cellular stresses including infection, trauma, and oxidative damage. However, if uncompensated, the UPR triggers apoptotic cell death. We demonstrate that the three signaling branches of UPR including the PERK, ATF6, and IRE1α are rapidly initiated in a mouse model of contusive SCI specifically at the injury epicenter. Immunohistochemical analyses of the various UPR markers revealed that in neurons, the UPR appeared at 6 and 24-h post-SCI. In contrast, in oligodendrocytes and astroglia, UPR persisted at least for up to 3 days post-SCI. The UPR-associated proapoptotic transcriptional regulator CHOP was among the UPR markers upregulated in neurons and oligodendrocytes, but not in astrocytes, of traumatized mouse spinal cords. To directly analyze its role in SCI, WT and CHOP null mice received a moderate T9 contusive injury. Deletion of CHOP led to an overall attenuation of the UPR after contusive SCI. Furthermore, analyses of hindlimb locomotion demonstrated a significant functional recovery that correlated with an increase in white-matter sparing, transcript levels of myelin basic protein, and Claudin 11 and decreased oligodendrocyte apoptosis in CHOP null mice in contrast to WT animals. Thus, our study provides evidence that the UPR contributes to oligodendrocyte loss after traumatic SCI.


Subject(s)
Endoplasmic Reticulum Stress/physiology , Recovery of Function/physiology , Spinal Cord Injuries/physiopathology , Up-Regulation/physiology , Activating Transcription Factor 4/metabolism , Analysis of Variance , Animals , Apoptosis/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Caspase 3/genetics , Caspase 3/metabolism , DNA-Binding Proteins/metabolism , Disease Models, Animal , Endoplasmic Reticulum Stress/genetics , Eukaryotic Initiation Factor-2/metabolism , Female , Locomotion/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin Basic Protein/genetics , Myelin Basic Protein/metabolism , Nerve Fibers, Myelinated/metabolism , Nerve Fibers, Myelinated/pathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Oligodendrocyte Transcription Factor 2 , Oligodendroglia/metabolism , Oligodendroglia/pathology , RNA, Messenger/metabolism , Regulatory Factor X Transcription Factors , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Time Factors , Transcription Factor CHOP/deficiency , Transcription Factors/metabolism , Unfolded Protein Response/genetics , Unfolded Protein Response/physiology , Up-Regulation/genetics
5.
Microcirculation ; 17(7): 557-67, 2010 Oct.
Article in English | MEDLINE | ID: mdl-21040121

ABSTRACT

UNLABELLED: We have demonstrated that MFs isolated from adipose retain angiogenic potential in vitro and form a mature, perfused network when implanted. However, adipose-derived microvessels are rich in provascularizing cells that could uniquely drive neovascularization in adipose-derived MFs implants. OBJECTIVE: Investigate the ability of MFs from a different vascular bed to recapitulate adipose-derived microvessel angiogenesis and network formation and analyze adipose-derived vessel plasticity by assessing whether vessel function could be modulated by astrocyte-like cells. METHODS: MFs were isolated by limited collagenase digestion from rodent brain or adipose and assembled into 3D collagen gels in the presence or absence of GRPs. The resulting neovasculatures that formed following implantation were assessed by measuring 3D vascularity and vessel permeability to small and large molecular tracers. RESULTS: Similar to adipose-derived MFs, brain-derived MFs can sprout and form a perfused neovascular network when implanted. Furthermore, when co-implanted in the constructs, GRPs caused adipose-derived vessels to express the brain endothelial marker glucose transporter-1 and to significantly reduce microvessel permeability. CONCLUSION: Neovascularization involving isolated microvessel elements is independent of the tissue origin and degree of vessel specialization. In addition, adipose-derived vessels have the ability to respond to environmental signals and change vessel characteristics.


Subject(s)
Microvessels/growth & development , Microvessels/transplantation , Neovascularization, Physiologic , Adipocytes/cytology , Adipocytes/transplantation , Animals , Astrocytes/cytology , Capillary Permeability , Cell Separation , Cerebral Cortex/blood supply , Embryonic Stem Cells/cytology , Embryonic Stem Cells/transplantation , Epididymis/blood supply , Epididymis/cytology , In Vitro Techniques , Male , Microvessels/cytology , Microvessels/physiology , Neuroglia/cytology , Neuroglia/transplantation , Rats
6.
Brain ; 133(Pt 4): 1026-42, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20375135

ABSTRACT

Blood vessel loss and inflammation cause secondary degeneration following spinal cord injury. Angiopoietin-1 through the Tie2 receptor, and other ligands through alphavbeta3 integrin, promote endothelial cell survival during developmental or tumour angiogenesis. Here, daily intravenous injections with an alphavbeta3-binding peptide named C16 or an angiopoietin-1 mimetic following a spinal cord contusion at thoracic level 9 in mice rescued epicentre blood vessels, white matter and locomotor function, and reduced detrimental inflammation. Preserved vascularity and reduced inflammation correlated with improved outcomes. C16 and angiopoietin-1 reduced leukocyte transmigration in vitro. Growth factor receptors and integrins facilitate each others' function. Therefore, angiopoietin-1 and C16 were combined and the effects were additive, resulting in almost complete functional recovery. The treatment had lasting effects when started 4 h following injury and terminated after one week. These results identify alphavbeta3 integrin and the endothelial-selective angiopoietin-1 as vascular and inflammatory regulators that can be targeted in a clinically relevant manner for neuroprotection after central nervous system trauma.


Subject(s)
Angiopoietin-1/administration & dosage , Integrin alphaVbeta3/administration & dosage , Neuroprotective Agents/administration & dosage , Peptide Fragments/administration & dosage , Spinal Cord Injuries/prevention & control , Spinal Cord/blood supply , Spinal Cord/drug effects , Amino Acid Sequence , Animals , Cell Movement/drug effects , Cell Movement/physiology , Drug Therapy, Combination , Female , Humans , Injections, Intravenous , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Recovery of Function/drug effects , Recovery of Function/physiology , Spinal Cord Injuries/physiopathology , Thoracic Vertebrae , Time Factors
7.
ASN Neuro ; 1(3)2009 Aug 26.
Article in English | MEDLINE | ID: mdl-19663807

ABSTRACT

Microvascular dysfunction, loss of vascular support, ischaemia and sub-acute vascular instability in surviving blood vessels contribute to secondary injury following SCI (spinal cord injury). Neither the precise temporal profile of the cellular dynamics of spinal microvasculature nor the potential molecular effectors regulating this plasticity are well understood. TGFß (transforming growth factor ß) isoforms have been shown to be rapidly increased in response to SCI and CNS (central nervous system) ischaemia, but no data exist regarding their contribution to microvascular dysfunction following SCI. To examine these issues, in the present study we used a model of focal spinal cord ischaemia/reperfusion SCI to examine the cellular response(s) of affected microvessels from 30 min to 14 days post-ischaemia. Spinal endothelial cells were isolated from affected tissue and subjected to focused microarray analysis of TGFß-responsive/related mRNAs 6 and 24 h post-SCI. Immunohistochemical analyses of histopathology show neuronal disruption/loss and astroglial regression from spinal microvessels by 3 h post-ischaemia, with complete dissolution of functional endfeet (loss of aquaporin-4) by 12 h post-ischaemia. Coincident with this microvascular plasticity, results from microarray analyses show 9 out of 22 TGFß-responsive mRNAs significantly up-regulated by 6 h post-ischaemia. Of these, serpine 1/PAI-1 (plasminogen-activator inhibitor 1) demonstrated the greatest increase (>40-fold). Furthermore, uPA (urokinase-type plasminogen activator), another member of the PAS (plasminogen activator system), was also significantly increased (>7.5-fold). These results, along with other select up-regulated mRNAs, were confirmed biochemically or immunohistochemically. Taken together, these results implicate TGFß as a potential molecular effector of the anatomical and functional plasticity of microvessels following SCI.


Subject(s)
Endothelial Cells/metabolism , Microvessels/physiology , Reperfusion Injury/metabolism , Spinal Cord Ischemia/metabolism , Transcriptional Activation/physiology , Transforming Growth Factor beta/biosynthesis , Animals , Endothelial Cells/pathology , Female , Microvessels/pathology , Neuronal Plasticity/physiology , RNA, Messenger/biosynthesis , Random Allocation , Rats , Rats, Sprague-Dawley , Reperfusion Injury/pathology , Spinal Cord Ischemia/pathology
8.
Curr Neurovasc Res ; 6(2): 124-31, 2009 May.
Article in English | MEDLINE | ID: mdl-19442162

ABSTRACT

Acute loss of spinal cord vascularity followed by an endogenous adaptive angiogenic response with concomitant microvascular dysfunction is a hallmark of traumatic spinal cord injury (SCI). Recently, the potent vasoactive factor vascular endothelial growth factor (VEGF) has received much attention as a putative therapeutic for the treatment of various neurodegenerative disorders, including SCI. Exogenous VEGF exerts both protective and destabilizing effects on microvascular elements and tissue following SCI but the role of endogenous VEGF is unclear. In the present study, we systemically applied a potent and well characterized soluble VEGF antagonist to adult C57Bl/6 mice post-SCI to elucidate the relative contribution of VEGF on the acute evolving microvascular response and its impact on functional recovery. While the VEGF Trap did not alter vascular density in the injury epicenter or penumbra, an overall increase in the number of Griffonia simplicifolia isolectin-B4 bound microvessels was observed, suggesting a VEGF-dependency to more subtle aspects of endothelial plasticity post-SCI. Neutralizing endogenous VEGF neither attenuated nor exacerbated chronic histopathology or functional recovery. These results support the idea that overall, endogenous VEGF is not neuroprotective or detrimental following traumatic SCI. Furthermore, they suggest that angiogenesis in traumatically injured spinal tissue is regulated by multiple effectors and is not limited by endogenous VEGF activation of affected spinal microvessels.


Subject(s)
Microvessels/drug effects , Nerve Regeneration/drug effects , Recovery of Function/drug effects , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Vascular Endothelial Growth Factor A/metabolism , Analysis of Variance , Animals , Disease Models, Animal , Female , Immunoglobulin G/therapeutic use , Lectins/drug effects , Mice , Mice, Inbred C57BL , Microvessels/pathology , Receptors, Vascular Endothelial Growth Factor , Recombinant Fusion Proteins/therapeutic use , Recovery of Function/physiology , Spinal Cord Injuries/drug therapy , Time Factors , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/immunology , Vascular Endothelial Growth Factor Receptor-2/therapeutic use
9.
J Comp Neurol ; 512(2): 243-55, 2009 Jan 10.
Article in English | MEDLINE | ID: mdl-19003792

ABSTRACT

Endothelial cell (EC) loss and subsequent angiogenesis occur over the first week after spinal cord injury (SCI). To identify molecular mechanisms that could be targeted with intravenous (i.v.) treatments, we determined whether transmembrane "a disintegrin and metalloprotease" (ADAM) proteins are expressed in ECs of the injured spinal cord. ADAMs bind to integrins, which are important for EC survival and angiogenesis. Female adult C57Bl/6 mice with a spinal cord contusion had progressively more ADAM8 (CD156) immunostaining in blood vessels and individual ECs between 1 and 28 days following injury. Uninjured spinal cords had little ADAM8 staining. The increase in ADAM8 mRNA and protein was confirmed in spinal cord lysates, and ADAM8 mRNA was present in FACS-enriched ECs. ADAM8 colocalized extensively and exclusively with the EC marker PECAM and also with i.v.-injected lectins. Intravenous isolectin B4 (IB4) labels a subpopulation of blood vessels at and within the injury epicenter 3-7 days after injury, coincident with angiogenesis. Both ADAM8 and the proliferation marker Ki-67 were present in IB4-positive microvessels. ADAM8-positive proliferating cells were seen at the leading end of IB4-positive blood vessels. Angiogenesis was confirmed by BrdU incorporation, binding of i.v.-injected nucleolin antibodies, and MT1-MMP immunostaining in a subset of blood vessels. These data suggest that ADAM8 is vascular selective and plays a role in proliferation and/or migration of ECs during angiogenesis following SCI.


Subject(s)
ADAM Proteins/metabolism , Antigens, CD/metabolism , Endothelial Cells/metabolism , Membrane Proteins/metabolism , Neovascularization, Physiologic/physiology , Spinal Cord Injuries/metabolism , Adult , Animals , Biomarkers/metabolism , Blood Vessels/anatomy & histology , Blood Vessels/metabolism , Endothelial Cells/cytology , Female , Humans , Mice , Mice, Inbred C57BL , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Spinal Cord/cytology , Spinal Cord/metabolism , Spinal Cord/pathology , Spinal Cord Injuries/pathology
10.
J Cereb Blood Flow Metab ; 28(11): 1771-85, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18612314

ABSTRACT

Microvascular dysfunction is a critical pathology that underlies the evolution of secondary injury mechanisms after traumatic spinal cord injury (SCI). However, little is known of the molecular regulation of endothelial cell (EC) plasticity observed acutely after injury. One reason for this is the relative lack of methods to quickly and efficiently obtain highly enriched spinal microvascular ECs for high-throughput molecular and biochemical analyses. Adult C57Bl/6 mice received an intravenous injection of fluorescein isothiocyanate (FITC)-conjugated Lycopersicon esculentum lectin, and FITC-lectin-bound spinal microvessels were greatly enriched by fluorescence-activated cell sorter (FACS) purification. This technique allows for rapid (<1.5 h postmortem) isolation of spinal cord microvascular ECs (smvECs). The results from cell counting, reverse-transcription polymerase chain reaction (RT-PCR), and western blot analyses show a high degree of EC enrichment at mRNA and protein levels. Furthermore, a focused EC biology microarray analysis identified multiple mRNAs dramatically increased in the EC compartment 24 h after SCI, which is a time point associated with the pathologic loss of spinal vasculature. These included thrombospondin-1, CCL5/RANTES, and urokinase plasminogen activator, suggesting they may represent targets for therapeutic intervention. Furthermore, these novel methodologic approaches will likely facilitate the discovery of molecular regulators of endothelial dysfunction in a variety of central nervous system (CNS) disorders including stroke and other neurodegenerative diseases having a vascular component.


Subject(s)
Endothelium, Vascular/physiopathology , Gene Expression Profiling , Microcirculation/physiology , RNA, Messenger/genetics , Spinal Cord Injuries/genetics , Spinal Cord Injuries/physiopathology , Spinal Cord/blood supply , Transcription, Genetic , Animals , Annexins/genetics , Endothelium, Vascular/pathology , Female , Fibrinolysin/genetics , Flow Cytometry , Gene Expression Regulation , Immunohistochemistry , Mice , Mice, Inbred C57BL , Microcirculation/pathology , Oligonucleotide Array Sequence Analysis , Plant Lectins , Reverse Transcriptase Polymerase Chain Reaction , Thrombospondins/genetics , Urokinase-Type Plasminogen Activator/genetics
11.
J Comp Neurol ; 507(1): 1031-52, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18092342

ABSTRACT

After traumatic spinal cord injury (SCI), disruption and plasticity of the microvasculature within injured spinal tissue contribute to the pathological cascades associated with the evolution of both primary and secondary injury. Conversely, preserved vascular function most likely results in tissue sparing and subsequent functional recovery. It has been difficult to identify subclasses of damaged or regenerating blood vessels at the cellular level. Here, adult mice received a single intravenous injection of the Griffonia simplicifolia isolectin B4 (IB4) at 1-28 days following a moderate thoracic (T9) contusion. Vascular binding of IB4 was maximally observed 7 days following injury, a time associated with multiple pathologic aspects of the intrinsic adaptive angiogenesis, with numbers of IB4 vascular profiles decreasing by 21 days postinjury. Quantitative assessment of IB4 binding shows that it occurs within the evolving lesion epicenter, with affected vessels expressing a temporally specific dysfunctional tight junctional phenotype as assessed by occludin, claudin-5, and ZO-1 immunoreactivities. Taken together, these results demonstrate that intravascular lectin delivery following SCI is a useful approach not only for observing the functional status of neovascular formation but also for definitively identifying specific subpopulations of reactive spinal microvascular elements.


Subject(s)
Mice, Inbred C57BL/physiology , Neovascularization, Physiologic/physiology , Plant Lectins , Spinal Cord Injuries/physiopathology , Spine/blood supply , Adaptation, Physiological/physiology , Age Factors , Animals , Capillaries/cytology , Capillaries/physiology , Capillaries/ultrastructure , Cell Division/physiology , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Endothelium, Vascular/ultrastructure , Female , Mice , Microscopy, Immunoelectron , Spinal Cord Injuries/pathology , Spine/pathology , Tight Junctions/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...