Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters











Publication year range
1.
Data Brief ; 49: 109280, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37600128

ABSTRACT

Per- and polyfluoroalkyl substances (PFAS) are widespread and highly persistent organic chemicals with adverse health effects. The US Environmental Protection Agency has issued health advisory limits of 70 ng/L for aqueous concentrations of PFOA + PFOS. In the Colorado Springs, Colorado (USA), metro area, the Widefield Aquifer (groundwater) and Fountain Creek Watershed (surface water) have been contaminated by PFAS from aqueous film-forming foams. Here we present the concentrations of selected linear and branched isomers of legacy PFAS found in surface water (n = 95), soil (n = 83), and sediment (n = 34) samples collected from several creeks of the Fountain Creek Watershed. Collected samples were prepared for high-performance liquid chromatography tandem mass spectrometry (LC/MS/MS) analysis via liquid/liquid extraction and/or solid phase extraction (SPE). This dataset includes the geographic locations of sampled creeks, LC/MS/MS instrumental conditions, method verification data including percent recovery to assess method accuracy and background contamination of PFAS in laboratory reagents and supplies, and determined concentrations of PFAS in water, soil, and sediment samples. These locations were surveyed monthly for a full year and provide a rich dataset to assess influence of sampling location, temporal variability in concentration, and overall contaminant persistence.

2.
BMC Cancer ; 21(1): 923, 2021 Aug 16.
Article in English | MEDLINE | ID: mdl-34399705

ABSTRACT

BACKGROUND: Gastric cancer (GC) is one of the leading causes of cancer-related deaths worldwide. Human epidermal growth factor receptor 2 (HER2) amplification occurs in approximately 13-23% of all GC cases and patients with HER2 overexpression exhibit a poor prognosis. Lapatinib, a dual EGFR/HER2 tyrosine kinase inhibitor, is an effective agent to treat HER2-amplified breast cancer but it failed in gastric cancer (GC) clinical trials. However, the molecular mechanism of lapatinib resistance in HER2-amplified GC is not well studied. METHODS: We employed an unbiased, genome-scale screening with pooled CRISPR library on HER2-amplified GC cell lines to identify genes that are associated with resistance to lapatinib. To validate the candidate genes, we applied in vitro and in vivo pharmacological tests to confirm the function of the target genes. RESULTS: We found that loss of function of CSK or PTEN conferred lapatinib resistance in HER2-amplified GC cell lines NCI-N87 and OE19, respectively. Moreover, PI3K and MAPK signaling was significantly increased in CSK or PTEN null cells. Furthermore, in vitro and in vivo pharmacological study has shown that lapatinib resistance by the loss of function of CSK or PTEN, could be overcome by lapatinib combined with the PI3K inhibitor copanlisib and MEK inhibitor trametinib. CONCLUSIONS: Our study suggests that loss-of-function mutations of CSK and PTEN cause lapatinib resistance by re-activating MAPK and PI3K pathways, and further proved these two pathways are druggable targets. Inhibiting the two pathways synergistically are effective to overcome lapatinib resistance in HER2-amplified GC. This study provides insights for understanding the resistant mechanism of HER2 targeted therapy and novel strategies that may ultimately overcome resistance or limited efficacy of lapatinib treatment for subset of HER2 amplified GC.


Subject(s)
Biomarkers, Tumor/metabolism , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Receptor, ErbB-2/metabolism , Stomach Neoplasms/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Female , Gene Expression Profiling , Humans , Lapatinib/administration & dosage , Mice , Mice, Inbred NOD , Mice, SCID , Pyridones/administration & dosage , Pyrimidines/administration & dosage , Pyrimidinones/administration & dosage , Quinazolines/administration & dosage , Receptor, ErbB-2/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Lab Anim (NY) ; 50(7): 185-195, 2021 07.
Article in English | MEDLINE | ID: mdl-34127866

ABSTRACT

Animal models play a critical role in establishing causal relationships between gut microbiota and disease. The laboratory mouse is widely used to study the role of microbes in various disorders; however, differences between mouse vendors, genetic lineages and husbandry protocols have been shown to contribute to variation in phenotypes and to non-reproducibility of experimental results. We sought to understand how gut microbiome profiles of mice vary by vendor, vendor production facility and health status upon receipt into an academic facility and how they change over 12 weeks in the new environment. C57BL/6 mice were sourced from two different production sites for each of three different vendors. Mice were shipped to an academic research vivarium, and fresh-catch stool samples were collected from mice immediately from the shipping box upon receipt, and again after 2, 6 and 12 weeks in the new facility. Substantial variation in bacterial proportional abundance was observed among mice from each vendor at the time of receipt, but shared microbes accounted for most sequence reads. Vendor-specific microbes were generally of low abundance. Microbial profiles of mice from all vendors exhibited shifts over time, highlighting the importance of environmental conditions on microbial dynamics. Our results emphasize the need for continued efforts to account for sources of variation in animal models and understand how they contribute to experimental reproducibility.


Subject(s)
Gastrointestinal Microbiome , Animals , Bacteria , Feces , Mice , Mice, Inbred C57BL , Reproducibility of Results
5.
Biol Proced Online ; 23(1): 1, 2021 Jan 04.
Article in English | MEDLINE | ID: mdl-33390162

ABSTRACT

BACKGROUND: Gastric cancer metastasis is a highly fatal disease with a five-year survival rate of less than 5%. One major obstacle in studying gastric cancer metastasis is the lack of faithful models available. The cancer xenograft mouse models are widely used to elucidate the mechanisms of cancer development and progression. Current procedures for creating cancer xenografts include both heterotopic (i.e., subcutaneous) and orthotopic transplantation methods. Compared to the heterotopic model, the orthotopic model has been shown to be the more clinically relevant design as it enables the development of cancer metastasis. Although there are several methods in use to develop the orthotopic gastric cancer model, there is not a model which uses various types of tumor materials, such as soft tissues, semi-liquid tissues, or culture derivatives, due to the technical challenges. Thus, developing the applicable orthotopic model which can utilize various tumor materials is essential. RESULTS: To overcome the known limitations of the current orthotopic gastric cancer models, such as exposure of tumor fragments to the neighboring organs or only using firm tissues for the orthotopic implantation, we have developed a new method allowing for the complete insertion of soft tissue fragments or homogeneously minced tissues into the stomach submucosa layer of the immunodeficient NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ (NSG) mouse. With this completely-closed transplantation method, tumors with various types of tissue may be used to establish orthotopic gastric cancer models without the risks of exposure to nearby organs or cell leakage. This surgical procedure was highly reproducible in generating forty-eight mouse models with a surgery success rate of 96% and tumor formation of 93%. Among four orthotopic patient-derived xenograft (PDX) models that we generated in this study, we verified that the occurrence of organotropic metastasis in either the liver or peritoneal cavity was the same as that of the donor patients. CONCLUSION: Here we describe a new protocol, step by step, for the establishment of orthotopic xenograft of gastric cancer. This novel technique will be able to increase the use of orthotopic models in broader applications for not only gastric cancer research but also any research related to the stomach microenvironment.

6.
Proc Natl Acad Sci U S A ; 117(49): 30907-30917, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33219121

ABSTRACT

Myostatin (MSTN) is a transforming growth factor-ß (TGF-ß) family member that normally acts to limit muscle growth. The function of MSTN is partially redundant with that of another TGF-ß family member, activin A. MSTN and activin A are capable of signaling through a complex of type II and type I receptors. Here, we investigated the roles of two type II receptors (ACVR2 and ACVR2B) and two type I receptors (ALK4 and ALK5) in the regulation of muscle mass by these ligands by genetically targeting these receptors either alone or in combination specifically in myofibers in mice. We show that targeting signaling in myofibers is sufficient to cause significant increases in muscle mass, showing that myofibers are the direct target for signaling by these ligands in the regulation of muscle growth. Moreover, we show that there is functional redundancy between the two type II receptors as well as between the two type I receptors and that all four type II/type I receptor combinations are utilized in vivo. Targeting signaling specifically in myofibers also led to reductions in overall body fat content and improved glucose metabolism in mice fed either regular chow or a high-fat diet, demonstrating that these metabolic effects are the result of enhanced muscling. We observed no effect, however, on either bone density or muscle regeneration in mice in which signaling was targeted in myofibers. The latter finding implies that MSTN likely signals to other cells, such as satellite cells, in addition to myofibers to regulate muscle homeostasis.


Subject(s)
Activin Receptors, Type II/metabolism , Activin Receptors, Type I/metabolism , Activins/metabolism , Muscle Development , Myostatin/metabolism , Animals , Mice, Inbred C57BL , Muscle Fibers, Skeletal/metabolism , Muscles/metabolism , Organ Size
7.
Proc Natl Acad Sci U S A ; 117(38): 23942-23951, 2020 09 22.
Article in English | MEDLINE | ID: mdl-32900939

ABSTRACT

Among the physiological consequences of extended spaceflight are loss of skeletal muscle and bone mass. One signaling pathway that plays an important role in maintaining muscle and bone homeostasis is that regulated by the secreted signaling proteins, myostatin (MSTN) and activin A. Here, we used both genetic and pharmacological approaches to investigate the effect of targeting MSTN/activin A signaling in mice that were sent to the International Space Station. Wild type mice lost significant muscle and bone mass during the 33 d spent in microgravity. Muscle weights of Mstn-/- mice, which are about twice those of wild type mice, were largely maintained during spaceflight. Systemic inhibition of MSTN/activin A signaling using a soluble form of the activin type IIB receptor (ACVR2B), which can bind each of these ligands, led to dramatic increases in both muscle and bone mass, with effects being comparable in ground and flight mice. Exposure to microgravity and treatment with the soluble receptor each led to alterations in numerous signaling pathways, which were reflected in changes in levels of key signaling components in the blood as well as their RNA expression levels in muscle and bone. These findings have implications for therapeutic strategies to combat the concomitant muscle and bone loss occurring in people afflicted with disuse atrophy on Earth as well as in astronauts in space, especially during prolonged missions.


Subject(s)
Activins/metabolism , Bone Resorption/metabolism , Muscle, Skeletal/metabolism , Myostatin , Space Flight , Activin Receptors, Type II/genetics , Activin Receptors, Type II/metabolism , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscular Atrophy/metabolism , Myostatin/genetics , Myostatin/metabolism , Signal Transduction
8.
J Vis Exp ; (157)2020 03 04.
Article in English | MEDLINE | ID: mdl-32202516

ABSTRACT

Human brain organoids differentiated from embryonic stem cells offer the unique opportunity to study complicated interactions of multiple cell types in a three-dimensional system. Here we present a relatively straightforward and inexpensive method that yields brain organoids. In this protocol human pluripotent stem cells are broken into small clusters instead of single cells and grown in basic media without a heterologous basement membrane matrix or exogenous growth factors, allowing the intrinsic developmental cues to shape the organoid's growth. This simple system produces a diversity of brain cell types including glial and microglial cells, stem cells, and neurons of the forebrain, midbrain, and hindbrain. Organoids generated from this protocol also display hallmarks of appropriate temporal and spatial organization demonstrated by brightfield images, histology, immunofluorescence and real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR). Because these organoids contain cell types from various parts of the brain, they can be utilized for studying a multitude of diseases. For example, in a recent paper we demonstrated the use of organoids generated from this protocol for studying the effects of hypoxia on the human brain. This approach can be used to investigate an array of otherwise difficult to study conditions such as neurodevelopmental handicaps, genetic disorders, and neurologic diseases.


Subject(s)
Human Embryonic Stem Cells/cytology , Organoids , Brain/cytology , Brain/metabolism , Cell Differentiation , Humans , Mesencephalon , Neurons/cytology , Organoids/metabolism , Pluripotent Stem Cells/cytology , Tissue Culture Techniques
9.
J Emerg Manag ; 18(6): 525-534, 2020.
Article in English | MEDLINE | ID: mdl-33428207

ABSTRACT

OBJECTIVE: The emergency operations center (EOC) is an essential component of modern emergency management. Traditionally understood as a place where officials communicate with the public, support coordination, manage operations, craft policy, gather information, and host visitors; there has been little recent research on their structure, operations, or work procedures. EOCs may in fact be, as we argue here, places where emergency managers come to find workarounds, delegate tasks, and find new sources of expertise in order to make sense, make meaning, and make decisions. However, despite their status as a symbol of emergency management and recipients of large amounts of funding, there has been relatively little scientific research into the EOC. With this paper, we synthesize the existing research and propose a variety of research questions to accelerate the process of inquiry into the EOC. DESIGN: Informed by an extensive literature review, this article presents a comprehensive look at the existing state of knowledge surrounding EOCs. INTERVENTIONS: Research questions to support investigation of the EOC are suggested. CONCLUSIONS: The EOC is an underexplored setting ripe for development and discovery by researchers and emergency managers seeking to influence the field of emergency management.


Subject(s)
Emergencies , Humans
10.
Ann Hematol ; 98(9): 2089-2096, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31203423

ABSTRACT

Myelodysplastic syndromes are clonal hematopoietic stem cell disorders characterized by cytopenia and intramedullary apoptosis. BCL-2 Ovarian Killer (BOK) is a pro-apoptotic member of the BCL-2 family of proteins which, when stabilized from endoplasmic reticulum-associated degradation (ERAD), induces apoptosis in response to ER stress. Although ER stress appropriately activates the unfolded protein response (UPR) in BOK-disrupted cells, the downstream effector signaling that includes ATF4 is defective. We used Nup98-HoxD13 (NHD13) transgenic mice to evaluate the consequences of BOK loss on hematopoiesis and leukemogenesis. Acute myeloid leukemia developed in 36.7% of NHD13 mice with a Bok gene knockout between the age of 8 and 13 months and presented a similar overall survival to the NHD13 mice. The loss of BOK exacerbated anemia in NHD13 mice, and NHD13/BOK-deficient mice exhibited significantly lower hemoglobin, lower mean cell hemoglobin concentration, and higher mean cell volume than NHD13 mice. Hematopoietic progenitor cell assays revealed a decreased amount of erythroid progenitor stem cells (BFU-E) in the bone marrow of NHD13-transgenic/BOK-deficient mice. RT-qPCR analysis demonstrated decreased mean value of ATF4 in the erythroid progenitors of NHD13 and NHD13/BOK-deficient mice. Our results suggest that in addition to induction of apoptosis in response to ER stress, BOK may regulate erythropoiesis when certain erythroid progenitors experience cell stress.


Subject(s)
Endoplasmic Reticulum Stress , Endoplasmic Reticulum-Associated Degradation , Erythroid Precursor Cells/metabolism , Erythropoiesis , Myelodysplastic Syndromes/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Apoptosis/genetics , Disease Models, Animal , Erythroid Precursor Cells/pathology , Hemoglobins/metabolism , Mice , Mice, Knockout , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Proto-Oncogene Proteins c-bcl-2/genetics
11.
Cell Death Dis ; 10(4): 325, 2019 04 11.
Article in English | MEDLINE | ID: mdl-30975982

ABSTRACT

Neonatal hypoxic injury (NHI) is a devastating cause of disease that affects >60% of babies born with a very low birth weight, resulting in significant morbidity and mortality, including life-long neurological consequences such as seizures, cerebral palsy, and intellectual disability. Hypoxic injury results in increased neuronal death, which disrupts normal brain development. Although animal model systems have been useful to study the effects of NHI, they do not fully represent the uniqueness and complexities of the human brain. To better understand the effects of hypoxia on human brain development, we have generated a brain organoid protocol and evaluated these cells over the course of 6 months. As anticipated, the expression of a forebrain marker, FOXG1, increased and then remained expressed over time, while there was a transition in the expression of the deep-layer (TBR1) and upper-layer (SATB2) cortical markers. In addition, ventral genes (Eng1 and Nkx2.1) as well as markers of specialized nonneuronal cells (Olig2 and GFAP) also increased at later time points. We next tested the development of our in vitro cerebral organoid model at different oxygen concentrations and found that hypoxia repressed gene markers for forebrain, oligodendrocytes, glial cells, and cortical layers, as well as genes important for the migration of cortical neurons. In contrast, ventral markers were either unaffected or even increased in expression with hypoxic insult. Interestingly, the negative effect of hypoxia on the dorsal brain genes as well as oligodendrocytes, and neuronal progenitors could be mitigated by the use of minocycline, an FDA-approved small molecule. Taken together, we have generated a unique and relevant in vitro human brain model system to study diseases such as NHI as well as their potential treatments. Using this system, we have shown the efficacy of minocycline for human NHI.


Subject(s)
Brain/metabolism , Hypoxia, Brain/drug therapy , Minocycline/therapeutic use , Cell Death/drug effects , Human Embryonic Stem Cells , Humans , Hypoxia, Brain/genetics , Hypoxia, Brain/metabolism , Hypoxia, Brain/prevention & control , Hypoxia-Ischemia, Brain/metabolism , Neurons/metabolism , Organoids/cytology , Organoids/drug effects , Organoids/metabolism , Organoids/pathology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Time Factors
13.
Behav Brain Res ; 333: 74-82, 2017 08 30.
Article in English | MEDLINE | ID: mdl-28666838

ABSTRACT

Matrix Metalloproteinase2, (MMP2, gelatinase A) is a zinc-containing enzyme with a broad substrate specificity including components of the extracellular matrix, cell surface molecules and a wide range bioactive molecules. MMP2 is known to play important roles in a variety of signaling pathways and processes in a wide range of cell types and tissues. In this report we elucidate the effects of the absence of MMP2 in Neural Precursor Cells (NPC) derived from C57BL/6 MMP2 KO mice and in primary and secondary neurosphere formation. We observed smaller neurosphere numbers and sizes, decreased NPC numbers, PCNA expression, DNA and Akt activation in MMP2 KO NPC compared to WT NPC. We also found decreased neurosphere formation and NPC migration outward from adherent neurospheres, decreased CXCR4 and nestin expression and increased GFAP and neuro-filament expression in MMP2 KO NPC compared to Wt NPC. MMP2 KO mice were found to exhibit increased anxiety manifested in open field activity assays compared to Wt mice. MMP2 KO mice also exhibited differences in motor activities manifested by decreased balance and endurance during Rota-rod testing. These studies illustrate an important role of MMP2 in cognitive and motor behaviors and confirm its importance in NPC activities crucial to brain development, growth and response to and recovery from injury.


Subject(s)
Cognition/physiology , Matrix Metalloproteinase 2/deficiency , Motor Activity/genetics , Neural Stem Cells/physiology , Animals , Animals, Newborn , Cell Movement/genetics , Cell Proliferation/genetics , Cells, Cultured , Exploratory Behavior/physiology , Gene Expression Regulation/genetics , Matrix Metalloproteinase 2/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/physiology , Nerve Tissue Proteins/metabolism , Neurogenesis/genetics , Oncogene Protein v-akt/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Receptors, CXCR4/metabolism , Spatial Learning/physiology
14.
Lab Invest ; 97(4): 356-369, 2017 04.
Article in English | MEDLINE | ID: mdl-28092362

ABSTRACT

Chronic sublethal hypoxia, a complication of premature birth, is associated with cognitive and motor handicaps. Responsiveness to and recovery from this hypoxic environment is dependent on induction of HIF-1 α in the cells affected. Microvascular endothelial-glial and microvascular endothelial-neuronal precursor interactions have been found to be dynamic and reciprocal, involving autocrine and paracrine signaling, with response and recovery correlated with baseline levels and levels of induction of HIF-1 α.To ascertain the roles of endothelial HIF-1 α in the responses of brain microvascular endothelial cells (EC) and neuronal precursors to hypoxia, we examined the effects of the presence and absence of endothelial HIF-1 α expression in culture and in cells comprising the subventricular zone (SVZ) and dentate gyrus under normoxic and hypoxic conditions. We used C57BL/6 WT and EC HIF-1 α -deficient mice and brain microvascular ECs isolated from these mice in western blots, immunofluorescence, and behavioral studies to examine the roles of EC HIF-1 α behaviors of endothelial and neuronal precursor cells (NPCs) in SVZ and hippocampal tissues under normoxic and hypoxic conditions and behaviors of these mice in open field activity tests. Analyses of ECs and SVZ and dentate gyrus tissues revealed effects of the absence of endothelial HIF-1 α on proliferation and apoptosis as well as open field activity, with both ECs and neuronal cells exhibiting decreased proliferation, increased apoptosis, and pups exhibiting gender-specific differences in open field activities. Our studies demonstrate the autocrine and paracrine effects of EC HIF-1 α-modulating proliferative and apoptotic behaviors of EC and NPC in neurogenic regions of the brain and gender-specific behaviors in normoxic and hypoxic settings.


Subject(s)
Dentate Gyrus/metabolism , Endothelial Cells/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lateral Ventricles/metabolism , Animals , Animals, Newborn , Apoptosis , Blotting, Western , Cell Hypoxia , Cell Proliferation , Cells, Cultured , Dentate Gyrus/cytology , Female , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lateral Ventricles/cytology , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence , Motor Activity , Neural Stem Cells/metabolism
16.
Proc Natl Acad Sci U S A ; 112(23): 7201-6, 2015 Jun 09.
Article in English | MEDLINE | ID: mdl-26015568

ABSTRACT

B-cell lymphoma 2 (BCL-2) ovarian killer (BOK) is a BCL-2 family protein with high homology to the multidomain proapoptotic proteins BAX and BAK, yet Bok(-/-) and even Bax(-/-)Bok(-/-) and Bak(-/-)Bok(-/-) mice were reported to have no overt phenotype or apoptotic defects in response to a host of classical stress stimuli. These surprising findings were interpreted to reflect functional compensation among the BAX, BAK, and BOK proteins. However, BOK cannot compensate for the severe apoptotic defects of Bax(-/-)Bak(-/-) mice despite its widespread expression. Here, we independently developed Bok(-/-) mice and found that Bok(-/-) cells are selectively defective in their response to endoplasmic reticulum (ER) stress stimuli, consistent with the predominant subcellular localization of BOK at the ER. Whereas Bok(-/-) mouse embryonic fibroblasts exposed to thapsigargin, A23187, brefeldin A, DTT, geldanamycin, or bortezomib manifested reduced activation of the mitochondrial apoptotic pathway, the death response to other stimuli such as etoposide, staurosporine, or UV remained fully intact. Multiple organs in Bok(-/-) mice exhibited resistance to thapsigargin-induced apoptosis in vivo. Although the ER stress agents activated the unfolded protein response, both ATF4 and CHOP activation were diminished in Bok(-/-) cells and mice. Importantly, BAX and BAK were unable to compensate for the defective apoptotic response to ER stress observed in SV40-transformed and primary Bok(-/-) cells, and in vivo. These findings support a selective and distinguishing role for BOK in regulating the apoptotic response to ER stress, revealing--to our knowledge--the first bona fide apoptotic defect linked to Bok deletion.


Subject(s)
Apoptosis/physiology , Endoplasmic Reticulum/metabolism , Oxidative Stress , Proto-Oncogene Proteins c-bcl-2/physiology , Activating Transcription Factor 4/metabolism , Animals , Annexin A5/pharmacology , Caspase 3/metabolism , Caspase 7/metabolism , Cells, Cultured , Endoplasmic Reticulum/enzymology , Enzyme Activation , Mice , Mice, Knockout , Proto-Oncogene Proteins c-bcl-2/genetics , Transcription Factor CHOP/metabolism
17.
Lab Invest ; 95(7): 765-80, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25961170

ABSTRACT

Microvascular endothelial cells cultured in three-dimensional hydrogel scaffolds form a network of microvessel structures when implanted subcutaneously in mice, inosculate with host vessels, and over time remodel into large ectatic vascular structures resembling hemangiomas. When compared with infantile hemangiomas, similarities were noted, including a temporal progression from a morphological appearance of a proliferative phase to the appearance of an involuted phase, mimicking the proliferative and involutional phases of infantile hemangioma. Consistent with the progression of a proliferative phase to an involuted phase, both the murine implants and human biopsy tissue exhibit reduced expression of Ajuba, YAP, and Survivin labeling as they progressed over time. Significant numbers of CD45+, CD11b+, Mac3+ mononuclear cells were found at the 2-week time point in our implant model that correlated with the presence of CD45+, CD68+ mononuclear cells observed in biopsies of human proliferative-phase hemangiomas. At the 4-week time point in our implant model, only small numbers of CD45+ cells were detected, which again correlated with our findings of significantly diminished CD45+, CD68+ mononuclear cells in human involutional-phase hemangiomas. The demonstration of mononuclear cell infiltration transiently in the proliferative phase of these lesions suggests that the vascular proliferation and/or regression may be driven in part by an immune response. Gross and microscopic morphological appearances of human proliferative and involutional hemangiomas and our implant model correlate well with each other as do the expression levels of Hippo pathway components (Ajuba and YAP) and Survivin and correlate with proliferation in these entities. Inhibitors of Survivin and Ajuba (which we have demonstrated to inhibit proliferation and increase apoptosis in murine hemangioendothelioma cell tissue culture) may have potential as other beneficial treatments for proliferating infantile hemangiomas. This implant model may have potential as a modest through-put screen for testing and development of therapeutics targeted at the proliferative phase of infantile hemangiomas, reducing the subsequent postinvolutional scarring or deformities sometimes associated with these lesions.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Disease Models, Animal , Hemangioma/metabolism , Inhibitor of Apoptosis Proteins/metabolism , LIM Domain Proteins/metabolism , Phosphoproteins/metabolism , Repressor Proteins/metabolism , Animals , Cell Cycle Proteins , Cells, Cultured , Child , Child, Preschool , Endothelial Cells/metabolism , Female , Hemangioma/immunology , Humans , Hydrogel, Polyethylene Glycol Dimethacrylate , Infant , Macrophages/metabolism , Male , Mice, Inbred C57BL , Survivin , Tissue Array Analysis , Tissue Scaffolds , YAP-Signaling Proteins
18.
PLoS One ; 8(10): e76265, 2013.
Article in English | MEDLINE | ID: mdl-24146847

ABSTRACT

Premature infants often experience chronic hypoxia, resulting in cognitive & motor neurodevelopmental handicaps. These sometimes devastating handicaps are thought to be caused by compromised neural precursor cell (NPC) repair/recovery resulting in variable central nervous system (CNS) repair/recovery. We have identified differential responses of two mouse strains (C57BL/6 & CD1) to chronic hypoxia that span the range of responsiveness noted in the premature human population. We previously correlated several CNS tissue and cellular behaviors with the different behavioral parameters manifested by these two strains. In this report, we use unbiased array technology to interrogate the transcriptome of the subventricular zone (SVZ) in these strains. Our results illustrate differences in mRNA expression in the SVZ of both C57BL/6 and CD1 mice following hypoxia as well as differences between C57BL/6 and CD1 SVZ under both normoxic and hypoxic conditions. Differences in expression were found in gene sets associated with Sox10-mediated neural functions that explain, in part, the differential cognitive and motor responsiveness to hypoxic insult. This may shed additional light on our understanding of the variable responses noted in the human premature infant population and facilitate early intervention approaches. Further interrogation of the differentially expressed gene sets will provide a more complete understanding of the differential responses to, and recovery from, hypoxic insult allowing for more informed modeling of the ranges of disease severity observed in the very premature human population.


Subject(s)
Cerebral Ventricles/blood supply , Cerebral Ventricles/metabolism , Gene Expression Profiling , Hypoxia/genetics , Hypoxia/pathology , Models, Neurological , Animals , Cerebral Ventricles/pathology , Databases, Genetic , Down-Regulation/genetics , Gene Ontology , Humans , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results , SOXE Transcription Factors/metabolism , Up-Regulation/genetics
19.
Cytokine Growth Factor Rev ; 24(4): 311-8, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23787159

ABSTRACT

Preclinical and clinical findings suggest that tumor-specific immune responses may be responsible--at least in part--for the clinical success of therapeutic regimens that rely on immunogenic cell death (ICD) inducers, including anthracyclines and oxaliplatin. The molecular pathways whereby some, but not all, cytotoxic agents promote bona fide ICD remain to be fully elucidated. Nevertheless, a central role for the endoplasmic reticulum (ER) stress response has been revealed in all scenarios of ICD described thus far. Hence, components of the ER stress machinery may constitute clinically relevant druggable targets for the induction of ICD. In this review, we will summarize recent findings in the field of ICD research with a special focus on ER stress mechanisms and their implication for cancer therapy.


Subject(s)
Cell Death/immunology , Endoplasmic Reticulum Stress/immunology , Endoplasmic Reticulum/immunology , Neoplasms/immunology , Animals , Anthracyclines/therapeutic use , Antineoplastic Agents/therapeutic use , Humans , Mice , Neoplasms/drug therapy , Neoplasms/radiotherapy , Organoplatinum Compounds/therapeutic use , Oxaliplatin
20.
Angiogenesis ; 16(3): 689-705, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23504212

ABSTRACT

Vascular integrity is a critical parameter in normal growth and development. Loss of appropriate vascular barrier function is present in various immune- and injury-mediated pathological conditions. CD44 is an adhesion molecule expressed by multiple cell types, including endothelial cells (EC). The goal of the present study was to examine how loss of CD44 affected vascular permeability. Using C57BL/6 WT and CD44-KO mice, we found no significant permeability to Evan's Blue in either strain at baseline. However, there was significantly increased histamine-induced permeability in CD44-deficient mice compared to WT counterparts. Similar results were observed in vitro, where CD44-deficient endothelial monolayers were also impermeable to 40kD-FITC dextran in the absence of vasoactive challenge, but exhibited enhanced and prolonged permeability following histamine. However, CD44-KO monolayers have reduced baseline barrier strength by electrical resistance, which correlated with increased permeability, at baseline, to smaller molecular weight 4-kD FITC-dextran, suggesting weakly formed endothelial junctions. The CD44-KO EC displayed several characteristics consistent with impaired barrier function/dysfunctional EC junctions, including differential expression, phosphorylation, and localization of endothelial junction proteins, increased matrix metalloprotease expression, and altered cellular morphology. Reduced platelet endothelial cell adhesion molecule-1 (PECAM-1) expression by CD44-KO EC in vivo and in vitro was also observed. Reconstitution of murine CD44 or PECAM-1 restored these defects to near WT status, suggesting CD44 regulates vascular permeability and integrity through a PECAM-1 dependent mechanism.


Subject(s)
Capillary Permeability/physiology , Endothelium, Vascular/physiology , Hyaluronan Receptors/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Animals , Blotting, Western , Capillary Permeability/genetics , Dextrans , Endothelium, Vascular/metabolism , Evans Blue , Fluorescein-5-isothiocyanate/analogs & derivatives , Hyaluronan Receptors/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL