Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
J Forensic Sci ; 68(4): 1310-1316, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37248532

ABSTRACT

A common requirement in the military, law enforcement, and forensic mission space is the need to collect trace samples from surfaces using a method that not only readily captures the sample but also retains its integrity for downstream identification and characterization. Additionally, collecting samples from three-dimensional objects (e.g., shell casings) is a challenge for which there is currently no validated standardized approach. Recently, hydrogels have been shown to have the potential for surface collection of trace bacterial spores, amino acids, and DNA. To test whether these hydrogels can serve as a viable collection medium for sampling DNA from surfaces, we carried out a series of preliminary tests examining collection efficiency and suitability of hydrogel material to recover samples of diluted, dried human DNA on a smooth polycarbonate surface. The recovery of surface DNA using a commercially available hydrogel was examined, and the efficiency compared to samples collected using a standard foam collection swab. DNA collected using the hydrogel and swab methods was then examined using quantitative polymerase chain reaction (qPCR) and short tandem repeat (STR) analysis to determine whether the collection material was compatible with these downstream processes. The hydrogel material used for this study collected the experimental DNA with comparable efficiency to standard collection swabs. In addition, qPCR and STR analyses demonstrated compatibility with the hydrogel collection and extraction process. These data suggest that hydrogels have the potential to be used as sample collection materials and deserve further characterization to elucidate their utility in collection from irregularly shaped, three-dimensional surfaces/materials.


Subject(s)
Hydrogels , Microsatellite Repeats , Humans , Specimen Handling/methods , DNA , DNA Fingerprinting
2.
PLoS One ; 18(2): e0280883, 2023.
Article in English | MEDLINE | ID: mdl-36780485

ABSTRACT

Organ-on-a-chip platforms are utilized in global bioanalytical and toxicological studies as a way to reduce materials and increase throughput as compared to in vivo based experiments. These platforms bridge the infrastructure and regulatory gaps between in vivo animal work and human systems, with models that exemplify active biological pathways. In conjunction with the advent of increased capabilities associated with next generation sequencing and mass spectrometry based '-omic' technologies, organ-on-a-chip platforms provide an excellent opportunity to investigate the global changes at multiple biological levels, including the transcriptome, proteome and metabolome. When investigated concurrently, a complete profile of cellular and regulatory perturbations can be characterized following treatment with specific agonists. In this study, global effects were observed and analyzed following liver chip exposure to the chemical warfare agent, VX. Even though the primary mechanism of action of VX (i.e. acetylcholinesterase inhibition) is well characterized, recent in vivo studies suggest additional protein binding partners that are implicated in metabolism and cellular energetic pathways. In addition, secondary toxicity associated with peripheral organ systems, especially in human tissues, is not well defined. Our results demonstrate the potential of utilizing an organ-on-a-chip platform as a surrogate system to traditional in vivo studies. This is realized by specifically indicating significant dysregulation of several cellular processes in response to VX exposure including but not limited to amino acid synthesis, drug metabolism, and energetics pathways.


Subject(s)
Chemical Warfare Agents , Animals , Humans , Chemical Warfare Agents/toxicity , Acetylcholinesterase , Microphysiological Systems , Multiomics
3.
Metabolites ; 12(9)2022 Aug 30.
Article in English | MEDLINE | ID: mdl-36144218

ABSTRACT

Sulfur mustard (HD) poses a serious threat due to its relatively simple production process. Exposure to HD in the short-term causes an inflammatory response, while long-term exposure results in DNA and RNA damage. Respiratory tract tissue models were exposed to relatively low concentrations of HD and collected at 3 and 24 h post exposure. Histology, cytokine ELISAs, and mass spectrometric-based analyses were performed. Histology and ELISA data confirmed previously seen lung damage and inflammatory markers from HD exposure. The multi-omic mass spectrometry data showed variation in proteins and metabolites associated with increased inflammation, as well as DNA and RNA damage. HD exposure causes DNA and RNA damage that results in variation of proteins and metabolites that are associated with transcription, translation and cellular energy.

4.
J Forensic Sci ; 66(2): 758-765, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33275295

ABSTRACT

The opioid crisis has continued to progress in the United States and the rest of the world. As this crisis continues, there is a pressing need for a rapid and cost-effective method for detecting fentanyl. Recent studies have suggested that lateral flow immunoassays (LFIs) could fill this technology gap. These qualitative paper-based assays contain antibodies designed to react with fentanyl and provide positive or negative results within a matter of minutes. In this study, two different LFI configurations for the detection of fentanyl were examined (dipsticks and cassettes) for effectiveness of detection using seized drug samples and postmortem urine samples. In the current study, 44 seized drug samples (32 fentanyl-positive, 12 fentanyl-negative) and 14 postmortem urine samples (10 fentanyl-positive, 4 fentanyl-negative) were analyzed. All 32 fentanyl-containing seized drug samples and 10 postmortem fentanyl-positive urine samples displayed positive LFI results with both LFI configurations. The fentanyl dipsticks displayed a sensitivity of 100%, a specificity of 75%, and an efficiency of 93.2% for seized drug samples and a sensitivity, specificity, and efficiency of 100% for postmortem urine. Analysis of the fentanyl cassettes displayed a sensitivity, specificity, and efficiency of 100% for seized drug samples and a sensitivity of 100%, a specificity of 75%, and an efficiency of 92.9% for postmortem urine samples. These data point to the utility of LFIs as a quick and low resource-dependent option for presumptive detection of fentanyl in real-world situations.


Subject(s)
Fentanyl/analysis , Illicit Drugs/analysis , Immunoassay/methods , Opioid-Related Disorders/urine , Gas Chromatography-Mass Spectrometry , Humans , Sensitivity and Specificity , Substance Abuse Detection
5.
J Forensic Sci ; 65(4): 1315-1323, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32017101

ABSTRACT

Following an intentional or accidental bio-warfare agent (BWA) release, environmental sample analysis is absolutely critical to determine the extent of contamination. When dealing with nonspore forming BWA (e.g., Yersinia pestis), retention of cell viability is central to such analyses. Even though significant advances have been achieved in DNA sequencing technologies, a positive identification of BWAs in environmental samples must be made through the ability of cells to form colony-forming units upon culturing. Inability to revive the cells between collection and analysis renders such studies inconclusive. Commercial kits designed to preserve the viability of pathogens contained within clinical samples are available, but many of them have not been examined for their ability to preserve samples containing suspected BWAs. The study was initiated to examine the applicability of commercial solutions aiding in retention of Y. pestis viability in samples stored under nonpermissive temperatures, that is, 40 and 37°C. While none of the tested solutions sustained cell viability at 40°C, the results show five out of 17 tested preservatives were capable of supporting viability of Y. pestis at 37°C.


Subject(s)
Cell Survival , Environmental Microbiology , Yersinia pestis/cytology , Yersinia pestis/genetics , Biological Warfare Agents , Cells, Cultured , Forensic Sciences , Humans , Polymerase Chain Reaction , Specimen Handling , Temperature
6.
Forensic Sci Int ; 300: 75-81, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31078080

ABSTRACT

In 2017, 47,600 overdose deaths were reported to be associated with the abuse of opioids, including prescription painkillers (e.g. oxycodone), opiates (e.g. heroin), or synthetic opioids (e.g. fentanyl) within the United States. The recent spike in the presence of synthetic opioids in lots of heroin distributed on the street present specific and significant challenges to law enforcement. Synthetic opioids are extremely toxic substances, which can easily be inhaled. This type of exposure can lead to accidental overdoses by law enforcement and other first responders answering calls involving illicit drugs containing these substances. Due to this extreme toxicity, it is important for these individuals to have tools that can be easily deployed for accurate presumptive field tests. Currently, there are only a limited number of presumptive tests available for fentanyl detection. In this study, we addressed this technology gap by evaluating newly developed lateral flow immunoassays (LFIs) designed for the detection of fentanyl and its derivatives. These LFIs were evaluated for effectiveness in different biofluid matrices, following an in vivo exposure, cross-reactivity with fentanyl analogs, and in case samples. This study demonstrates that LFIs have the potential to be used by law enforcement for the detection of synthetic opioids.


Subject(s)
Analgesics, Opioid/analysis , Fentanyl/analysis , Illicit Drugs/analysis , Immunoassay/methods , Animals , Fentanyl/analogs & derivatives , Humans , Limit of Detection , Opioid-Related Disorders/diagnosis , Rabbits , Saliva/chemistry , Substance Abuse Detection/methods
7.
J Forensic Sci ; 63(2): 412-419, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28585764

ABSTRACT

Environmental surface sampling is crucial in determining the zones of contamination and overall threat assessment. Viability retention of sampled material is central to such assessments. A systematic study was completed to determine viability of vegetative cells under nonpermissive storage conditions. Despite major gains in nucleic acid sequencing technologies, initial positive identification of threats must be made through direct culture of the sampled material using classical microbiological methods. Solutions have been developed to preserve the viability of pathogens contained within clinical samples, but many have not been examined for their ability to preserve biological agents. The purpose of this study was to systematically examine existing preservation materials that can retain the viability of Bacillus anthracis vegetative cells stored under nonpermissive temperatures. The results show effectiveness of five of seventeen solutions, which are capable of retaining viability of a sporulation deficient strain of B. anthracis Sterne when stored under nonrefrigerated conditions.


Subject(s)
Bacillus anthracis/cytology , Specimen Handling/instrumentation , Cell Survival , Forensic Sciences , Humans , Polymerase Chain Reaction , Temperature
8.
Chem Biol Interact ; 266: 38-46, 2017 Mar 25.
Article in English | MEDLINE | ID: mdl-28174098

ABSTRACT

Mesenchymal stem cells (MSCs) are multipotent cells located within various adult tissues. Recent literature has reported that human bone marrow-derived MSCs express active acetylcholinesterase (AChE) and that disruption of AChE activity by organophosphate (OP) chemicals decreases the ability of MSCs to differentiate into osteoblasts. The potential role of AChE in regulating MSC proliferation and differentiation is currently unknown. In the present study, we demonstrate that MSCs exposed to OPs have both decreased AChE activity and abundance. In addition, exposure to these OPs induced cellular death while decreasing cellular proliferation. Exposures to these compounds also reduced the adipogenic/osteogenic differentiation potentials of the MSCs. To elucidate the possible role of AChE in MSCs signaling following OP exposure, we captured potential AChE binding partners by performing polyhistidine (His8)-tagged AChE pulldowns, followed by protein identification using liquid chromatography mass spectrometry (LC-MS). Using this method, we determined that the focal adhesion protein, vinculin, is a potential binding partner with AChE in MSCs and these initial findings were confirmed with follow-up co-immunoprecipitation experiments. Identifying AChE binding partners helps to determine potential pathways associated with MSC proliferation and differentiation, and this understanding could lead to the development of future MSC-based tissue repair therapies.


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , Mesenchymal Stem Cells/drug effects , Organophosphates/pharmacology , Humans , Mesenchymal Stem Cells/cytology
9.
Int J Toxicol ; 34(5): 433-41, 2015.
Article in English | MEDLINE | ID: mdl-26173615

ABSTRACT

Organophosphorus (OP) pesticides are known to induce pulmonary toxicity in both humans and experimental animals. To elucidate the mechanism of OP-induced cytotoxicity, we examined the effects of parathion and malathion and their respective metabolites, paraoxon and malaoxon, on primary cultured human large and small airway cells. Exposure to paraoxon and malaoxon produced a dose-dependent increase in cytotoxicity following a 24-hour exposure, while treatment with parathion or malathion produced no effects at clinically relevant concentrations. Exposure to paraoxon-induced caspase activation, but malaoxon failed to induce this response. Since caspases have a major role in the regulation of apoptosis and cell death, we evaluated OP-induced cell death in the presence of a caspase inhibitor. Pharmacological caspase inhibition protected against paraoxon-induced cell death but not malaoxon-induced cell death. These data suggest that caspase activation is a key signaling element in paraoxon-induced cell death, but not malaoxon-induced cellular death in the pulmonary epithelium.


Subject(s)
Cholinesterase Inhibitors/toxicity , Epithelial Cells/drug effects , Insecticides/toxicity , Malathion/analogs & derivatives , Paraoxon/toxicity , Amino Acid Chloromethyl Ketones/pharmacology , Caspase Inhibitors/pharmacology , Caspases/metabolism , Cell Death/drug effects , Cell Survival/drug effects , Cells, Cultured , Epithelial Cells/metabolism , Humans , Malathion/toxicity , Parathion/toxicity , Respiratory System/cytology
10.
Am J Respir Cell Mol Biol ; 53(3): 303-13, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25569618

ABSTRACT

Resistin-like molecule α (RELMα) has mitogenic, angiogenic, vasoconstrictive, and chemokine-like properties and is highly relevant in lung pathology. Here, we used RELMα knockout (Retnla(-/-)) mice to investigate the role of RELMα in pulmonary vascular remodeling after intermittent ovalbumin (OVA) challenge. We compared saline- and OVA-exposed wild-type (WT) mice and found that OVA induced significant increases in right ventricular systolic pressure, cardiac hypertrophy, pulmonary vascular remodeling of intra-alveolar arteries, goblet cell hyperplasia in airway epithelium, and intensive lung inflammation, especially perivascular inflammation. Genetic ablation of Retnla prevented the OVA-induced increase in pulmonary pressure and cardiac hypertrophy seen in WT mice. Histological analysis showed that Retnla(-/-) mice exhibited less vessel muscularization, less perivascular inflammation, reduced medial thickness of intra-alveolar vessels, and fewer goblet cells in upper airway epithelium (250-600 µm) than did WT animals after OVA challenge. Gene expression profiles showed that genes associated with vascular remodeling, including those related to muscle protein, contractile fibers, and actin cytoskeleton, were expressed at a lower level in OVA-challenged Retnla(-/-) mice than in similarly treated WT mice. In addition, bronchoalveolar lavage from OVA-challenged Retnla(-/-) mice had lower levels of cytokines, such as IL-1ß, -1 receptor antagonist, and -16, chemokine (C-X-C motif) ligand 1, -2, -9, -10, and -13, monocyte chemoattractant protein-1, macrophage colony-stimulating factor, TIMP metallopeptidase inhibitor-1, and triggering receptor expressed on myeloid cells-1, than did that from WT mice when analyzed by cytokine array dot blots. Retnla knockout inhibited the OVA-induced T helper 17 response but not the T helper 2 response. Altogether, our results suggest that RELMα is involved in immune response-induced pulmonary vascular remodeling and the associated increase in inflammation typically observed after OVA challenge.


Subject(s)
Hypertension, Pulmonary/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Vascular Remodeling/immunology , Allergens/immunology , Animals , Cytokines/metabolism , Hypertension, Pulmonary/immunology , Hypertension, Pulmonary/physiopathology , Intercellular Signaling Peptides and Proteins/genetics , Lung/immunology , Lung/metabolism , Male , Mice, Inbred BALB C , Mice, Knockout , Ovalbumin/immunology
11.
Respir Res ; 14: 1, 2013 Jan 04.
Article in English | MEDLINE | ID: mdl-23289668

ABSTRACT

BACKGROUND: Both chronic hypoxia and allergic inflammation induce vascular remodeling in the lung, but only chronic hypoxia appears to cause PH. We investigate the nature of the vascular remodeling and the expression and role of hypoxia-induced mitogenic factor (HIMF/FIZZ1/RELMα) in explaining this differential response. METHODS: We induced pulmonary vascular remodeling through either chronic hypoxia or antigen sensitization and challenge. Mice were evaluated for markers of PH and pulmonary vascular remodeling throughout the lung vascular bed as well as HIMF expression and genomic analysis of whole lung. RESULTS: Chronic hypoxia increased both mean pulmonary artery pressure (mPAP) and right ventricular (RV) hypertrophy; these changes were associated with increased muscularization and thickening of small pulmonary vessels throughout the lung vascular bed. Allergic inflammation, by contrast, had minimal effect on mPAP and produced no RV hypertrophy. Only peribronchial vessels were significantly thickened, and vessels within the lung periphery did not become muscularized. Genomic analysis revealed that HIMF was the most consistently upregulated gene in the lungs following both chronic hypoxia and antigen challenge. HIMF was upregulated in the airway epithelial and inflammatory cells in both models, but only chronic hypoxia induced HIMF upregulation in vascular tissue. CONCLUSIONS: The results show that pulmonary vascular remodeling in mice induced by chronic hypoxia or antigen challenge is associated with marked increases in HIMF expression. The lack of HIMF expression in the vasculature of the lung and no vascular remodeling in the peripheral resistance vessels of the lung is likely to account for the failure to develop PH in the allergic inflammation model.


Subject(s)
Antigens , Hypertension, Pulmonary/etiology , Hypoxia/complications , Intercellular Signaling Peptides and Proteins/metabolism , Pneumonia/complications , Pulmonary Artery/metabolism , Th2 Cells/immunology , Animals , Arterial Pressure , Aspergillus/immunology , Chronic Disease , Disease Models, Animal , Familial Primary Pulmonary Hypertension , Gene Expression Profiling , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/immunology , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/etiology , Hypertrophy, Right Ventricular/immunology , Hypertrophy, Right Ventricular/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Ovalbumin/immunology , Pneumonia/immunology , Pneumonia/pathology , Pulmonary Artery/immunology , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Up-Regulation
12.
Inhal Toxicol ; 25(1): 37-62, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23293972

ABSTRACT

Chemical warfare agents (CWAs) as well as biological toxins present a significant inhalation injury risk to both deployed warfighters and civilian targets of terrorist attacks. Inhalation of many CWAs and biological toxins can induce severe pulmonary toxicity leading to the development of acute lung injury (ALI) as well as acute respiratory distress syndrome (ARDS). The therapeutic options currently used to treat these conditions are very limited and mortality rates remain high. Recent evidence suggests that human stem cells may provide significant therapeutic options for ALI and ARDS in the near future. The threat posed by CWAs and biological toxins for both civilian populations and military personnel is growing, thus understanding the mechanisms of toxicity and potential therapies is critical. This review will outline the pulmonary toxic effects of some of the most common CWAs and biological toxins as well as the potential role of stem cells in treating these types of toxic lung injuries.


Subject(s)
Biological Warfare Agents , Chemical Warfare Agents/toxicity , Lung/drug effects , Respiratory Distress Syndrome/therapy , Stem Cell Transplantation , Stem Cells , Toxins, Biological/toxicity , Animals , Humans , Lung/metabolism , Lung/pathology , Respiratory Distress Syndrome/chemically induced , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/pathology , Stem Cells/cytology , Stem Cells/physiology
13.
Stem Cells Dev ; 22(2): 239-47, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-22891677

ABSTRACT

Resistin-like molecule α (RELMα) is highly upregulated in the lungs of mice subjected to hypoxia. It is secreted from pulmonary epithelium and causes potent mitogenic, angiogenic, and vasoconstrictive effects in the lung vasculature. By using bone marrow transplantation in mice, we previously showed that RELMα is able to increase the number of bone marrow-derived cells in lung tissue, especially in the remodeling pulmonary vasculature. The current study investigated the effect of RELMα on progenitor stem cell content in mouse lung. Hypoxia, while stimulating RELMα expression, caused an increase in the number of Sca1(+)/CD45(-) progenitor cells in lungs of wild-type mice, but not in lungs of RELMα knockout mice. An in vitro study with cultured mesenchymal stem cells (MSCs) showed that RELMα induced a robust proliferative response that was dependent on Phosphatidylinositol 3-kinase/Akt and Erk activation. RELMα treatment of MSCs caused upregulation of a large number of genes involved in cell cycle, mitosis, organelle, and cytoskeleton biogenesis, and DNA metabolism. MSCs cultured in RELMα-supplemented media were able to maintain their differentiation potential into adipogenic, osteogenic, or mesenchymal phenotypes, although adipogenic differentiation was partially inhibited. These results demonstrate that RELMα may be involved in stem cell proliferation in the lung, without affecting differentiation potential.


Subject(s)
Cell Proliferation/drug effects , Intercellular Signaling Peptides and Proteins/metabolism , Mesenchymal Stem Cells/drug effects , Animals , Apoptosis , Bone Marrow/metabolism , Cell Count , Cell Differentiation/drug effects , Cell Division , Culture Media/metabolism , Female , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Hypoxia/metabolism , Hypoxia/pathology , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/pharmacology , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/metabolism , Lung/metabolism , Lung/pathology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Osteogenesis/drug effects , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
14.
Mol Cell Proteomics ; 10(3): M110.000901, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21139050

ABSTRACT

Hypoxia-induced mitogenic factor (HIMF) is a newly discovered protein that is up-regulated in murine models of pulmonary arterial hypertension and asthma. Our previous study shows that HIMF is a potent mitogenic, angiogenic, and vasoconstrictive chemokine associated with pulmonary arterial hypertension. Two-dimensional gel electrophoresis was used to investigate downstream molecules in HIMF-induced cell signaling, demonstrating that S100A11, an EF-hand calcium-binding protein, was exclusively altered and was decreased (2.7±0.2-fold, p<0.05) in pulmonary artery smooth muscle cells (SMCs) treated with HIMF for 5 min compared with untreated cells (n=4). Immunofluorescence showed that in control cells S100A11 is a cytosolic protein, which then aggregates and translocates both to the plasma membrane with subsequent exocytosis and to the nucleus upon HIMF stimulation. Annexin A2, a known S100A11 binding partner, also colocalized with S100A11 during HIMF-induced membrane trafficking. To investigate the intracellular function of S100A11, siRNA was used to knock down S100A11 expression in SMCs. The S100A11 knockdown significantly reduced HIMF-induced SMC migration but did not affect the SMC mitogenic action of HIMF. Our data show that S100A11 mediates HIMF-induced smooth muscle cell migration, vesicular exocytosis, and nuclear activation.


Subject(s)
Cell Movement/drug effects , Cell Nucleus/metabolism , Exocytosis/drug effects , Intercellular Signaling Peptides and Proteins/pharmacology , Myocytes, Smooth Muscle/cytology , S100 Proteins/metabolism , Secretory Vesicles/metabolism , Annexin A2/metabolism , Calcium/metabolism , Cell Nucleus/drug effects , Cell Proliferation/drug effects , Down-Regulation/drug effects , Extracellular Space/drug effects , Extracellular Space/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Protein Transport/drug effects , Pulmonary Artery/cytology , Secretory Vesicles/drug effects , Signal Transduction/drug effects
15.
J Immunol ; 185(9): 5539-48, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20889544

ABSTRACT

Hypoxia-induced mitogenic factor (HIMF), also known as found in inflammatory zone 1 and resistin-like molecule α, belongs to a novel class of cysteine-rich secreted proteins. It exhibits mitogenic and chemotactic properties during pulmonary hypertension-associated vascular remodeling, as well as fibrogenic properties during pulmonary fibrosis. HIMF expression in the lung was reported to be regulated by Th2 cytokines (IL-4 and IL-13) via the transcription factor STAT6 pathway in a bleomycin-induced pulmonary fibrosis model. However, in this study, we found that in the hypoxia-induced pulmonary hypertension model, lung HIMF expression is increased in IL-4 and STAT6 knockout (KO) mice to the same degree as in wild-type (WT) mice, suggesting that induction of HIMF expression does not require Th2 regulation in this model. We also found that HIMF-induced proliferative activity, hypertrophy, collagen, and extracellular matrix deposition in the pulmonary arteries are significantly less in IL-4 KO mice than in WT mice. In addition, HIMF-induced production of angiogenic factors/chemokines, such as vascular endothelial growth factor, MCP-1, and stromal-derived factor-1, in the lung resident cells, as well as macrophage infiltration, were significantly suppressed in the lungs of IL-4 KO mice. We also show that IL-4 was significantly increased in the lungs of HIMF-treated WT mice. Our in vitro studies using pulmonary microvascular endothelial cells revealed that HIMF stimulated cell proliferation, vascular endothelial growth factor expression, and MCP-1 production in a manner that is dependent on the IL-4/IL-4Rα system. These findings suggest that IL-4 signaling may play a significant role in HIMF-induced lung inflammation and vascular remodeling.


Subject(s)
Endothelial Cells/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Interleukin-4/metabolism , Pneumonia/metabolism , Signal Transduction/immunology , Animals , Cell Movement , Cell Proliferation , Endothelial Cells/immunology , Enzyme-Linked Immunosorbent Assay , Extracellular Matrix/metabolism , Gene Expression , Gene Expression Regulation , Hypertension, Pulmonary/metabolism , Immunoblotting , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/immunology , Interleukin-4/immunology , Lung/blood supply , Lung/immunology , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Culture Techniques , Pneumonia/immunology , Pneumonia/pathology , Pulmonary Fibrosis/metabolism , Reverse Transcriptase Polymerase Chain Reaction
16.
PLoS One ; 5(6): e11251, 2010 Jun 22.
Article in English | MEDLINE | ID: mdl-20582166

ABSTRACT

BACKGROUND: Pulmonary hypertension (PH) is a disease of multiple etiologies with several common pathological features, including inflammation and pulmonary vascular remodeling. Recent evidence has suggested a potential role for the recruitment of bone marrow-derived (BMD) progenitor cells to this remodeling process. We recently demonstrated that hypoxia-induced mitogenic factor (HIMF/FIZZ1/RELM alpha) is chemotactic to murine bone marrow cells in vitro and involved in pulmonary vascular remodeling in vivo. METHODOLOGY/PRINCIPAL FINDINGS: We used a mouse bone marrow transplant model in which lethally irradiated mice were rescued with bone marrow transplanted from green fluorescent protein (GFP)(+) transgenic mice to determine the role of HIMF in recruiting BMD cells to the lung vasculature during PH development. Exposure to chronic hypoxia and pulmonary gene transfer of HIMF were used to induce PH. Both models resulted in markedly increased numbers of BMD cells in and around the pulmonary vasculature; in several neomuscularized small (approximately 20 microm) capillary-like vessels, an entirely new medial wall was made up of these cells. We found these GFP(+) BMD cells to be positive for stem cell antigen-1 and c-kit, but negative for CD31 and CD34. Several of the GFP(+) cells that localized to the pulmonary vasculature were alpha-smooth muscle actin(+) and localized to the media layer of the vessels. This finding suggests that these cells are of mesenchymal origin and differentiate toward myofibroblast and vascular smooth muscle. Structural location in the media of small vessels suggests a functional role in the lung vasculature. To examine a potential mechanism for HIMF-dependent recruitment of mesenchymal stem cells to the pulmonary vasculature, we performed a cell migration assay using cultured human mesenchymal stem cells (HMSCs). The addition of recombinant HIMF induced migration of HMSCs in a phosphoinosotide-3-kinase-dependent manner. CONCLUSIONS/SIGNIFICANCE: These results demonstrate HIMF-dependent recruitment of BMD mesenchymal-like cells to the remodeling pulmonary vasculature.


Subject(s)
Blood Vessels/cytology , Bone Marrow Cells/cytology , Hypoxia/physiopathology , Intercellular Signaling Peptides and Proteins/physiology , Lung/blood supply , Animals , Blotting, Western , Bone Marrow Transplantation , Chemotaxis , Dependovirus/genetics , Female , Genetic Vectors , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence
17.
Am J Physiol Lung Cell Mol Physiol ; 297(2): L263-70, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19429774

ABSTRACT

Hypoxia-induced mitogenic factor (HIMF), also known as "found in inflammatory zone 1" (FIZZ1) or resistin-like molecule-alpha (RELMalpha), is a profound vasoconstrictor of the pulmonary circulation and a strong mitogenic factor in pulmonary vascular smooth muscle. To further understand the mechanism of these contractile and mitogenic responses, we examined the effect of HIMF on intracellular Ca(2+) in human pulmonary artery smooth muscle cells (SMC). Ca(2+) imaging in fluo 4-loaded human pulmonary artery SMC revealed that recombinant murine HIMF increased intracellular Ca(2+) concentration ([Ca(2+)](i)) in a sustained and oscillatory manner. This increase occurred independent of extracellular Ca(2+) influx. Pretreatment of human pulmonary artery SMC with U-73122, a specific inhibitor of phosphatidylinositol-phospholipase C (PLC) completely prevented the HIMF-induced Ca(2+) signal. The [Ca(2+)](i) increase was also abolished by pretreatment with 2-aminoethoxydiphenyl borate (2-APB), an inositol 1,4,5-trisphosphate (IP(3)) receptor antagonist. Ryanodine pretreatment did not affect initiation of [Ca(2+)](i) activation or internal release but reduced [Ca(2+)](i) at the plateau phase. Pretreatment with the Galpha(i)-specific inhibitor pertussis toxin and the Galpha(s)-specific inhibitor NF-449 did not block the Ca(2+) signal. Knockdown of Galpha(q/11) expression did not prevent Ca(2+) release, but the pattern of Ca(2+) release changed from the sustained oscillatory transients with prolonged plateau to a series of short [Ca(2+)](i) transients that return to baseline. However, pretreatment with the tyrosine kinase inhibitor genistein completely inhibited the internal Ca(2+) release. These results demonstrate that HIMF can stimulate intracellular Ca(2+) release in human pulmonary artery SMC through the PLC signaling pathway in an IP(3)- and tyrosine phosphorylation-dependent manner and that Galpha(q/11) protein-coupled receptor and ryanodine receptor contribute to the increase of [Ca(2+)](i).


Subject(s)
Calcium/metabolism , Hypoxia/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Pulmonary Artery/metabolism , Type C Phospholipases/metabolism , Aniline Compounds , Calcium Signaling/physiology , Estrenes/pharmacology , Fluorescent Dyes , Heterotrimeric GTP-Binding Proteins/genetics , Heterotrimeric GTP-Binding Proteins/metabolism , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Phosphodiesterase Inhibitors/pharmacology , Phosphorylation/physiology , Pulmonary Artery/cytology , Pyrrolidinones/pharmacology , RNA, Small Interfering , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Type C Phospholipases/antagonists & inhibitors , Tyrosine/metabolism , Vasoconstriction/physiology , Xanthenes
18.
J Immunol ; 182(9): 5469-76, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19380795

ABSTRACT

IL-4-mediated proangiogenic and proinflammatory vascular responses have been implicated in the pathogenesis of chronic lung diseases such as asthma. Although it is well known that hypoxia induces pulmonary angiogenesis and vascular alterations, the underlying mechanism of IL-4 on the pulmonary vasculature under hypoxic conditions remains unknown. In this context, we designed the present study to determine the functional importance of IL-4 for pulmonary angiogenesis under hypoxic conditions using IL-4 knockout (KO) animals. Our results show that hypoxia significantly increased IL-4R alpha expression in wild-type (WT) control lungs. Even though hypoxia significantly up-regulated vascular endothelial growth factor (VEGF) receptor expression in the lungs of both genotypes, hypoxia-induced VEGF, VCAM-1, HIF-1alpha, and ERK phosphorylation were significantly diminished in IL-4 KO lungs as compared with WT control lungs. In addition, hypoxia-induced pulmonary angiogenesis and proliferating activities in the airway and pulmonary artery were significantly suppressed in IL-4 KO lungs as compared with WT control lungs. We also isolated primary lung fibroblasts from these genotypes and stimulated these cells with hypoxia. Hypoxia-induced VEGF production was significantly suppressed in lung fibroblasts from IL-4 KO mice. These in vitro results are in accordance with the in vivo data. Furthermore, we observed a significant increase of hypoxia-induced pulmonary angiogenesis in STAT6 KO mice similar to that in WT controls. In conclusion, IL-4 has proangiogenic properties in the lung under hypoxic conditions via the VEGF pathway, and this is independent of the STAT6 pathway.


Subject(s)
Angiogenic Proteins/physiology , Hypoxia/immunology , Interleukin-4/physiology , Lung/immunology , Lung/metabolism , Neovascularization, Physiologic/immunology , Angiogenic Proteins/deficiency , Angiogenic Proteins/genetics , Animals , Cells, Cultured , Chronic Disease , Hypoxia/pathology , Hypoxia/physiopathology , Inflammation Mediators/metabolism , Inflammation Mediators/physiology , Interleukin-4/deficiency , Interleukin-4/genetics , Lung/blood supply , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Physiologic/genetics , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Receptors, Vascular Endothelial Growth Factor/biosynthesis , Receptors, Vascular Endothelial Growth Factor/genetics , STAT6 Transcription Factor/deficiency , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/physiology , Signal Transduction/genetics , Signal Transduction/immunology , Up-Regulation/genetics , Up-Regulation/immunology
19.
Am J Respir Cell Mol Biol ; 41(5): 553-61, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19251945

ABSTRACT

Scleroderma is a systemic, mixed connective tissue disease that can impact the lungs through pulmonary fibrosis, vascular remodeling, and the development of pulmonary hypertension and right heart failure. Currently, little is known about the molecular mechanisms that drive this condition, but we have recently identified a novel gene product that is up-regulated in a murine model of hypoxia-induced pulmonary hypertension. This molecule, known as hypoxia-induced mitogenic factor (HIMF), is a member of the newly described resistin gene family. We have demonstrated that HIMF has mitogenic, angiogenic, vasoconstrictive, inflammatory, and chemokine-like properties, all of which are associated with vascular remodeling in the lung. Here, we demonstrate that the human homolog of HIMF, resistin-like molecule (RELM)-beta, is expressed in the lung tissue of patients with scleroderma-associated pulmonary hypertension and is up-regulated compared with normal control subjects. Immunofluorescence colocalization revealed that RELM-beta is expressed in the endothelium and vascular smooth muscle of remodeled vessels, as well as in plexiform lesions, macrophages, T cells, and myofibroblast-like cells. We also show that addition of recombinant RELM-beta induces proliferation and activation of ERK1/2 in primary cultured human pulmonary endothelial and smooth muscle cells. These results suggest that RELM-beta may be involved in the development of scleroderma-associated pulmonary hypertension.


Subject(s)
Hypertension, Pulmonary/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Lung/metabolism , Pulmonary Fibrosis/metabolism , Scleroderma, Systemic/metabolism , Adult , Case-Control Studies , Cell Proliferation , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/pathology , Enzyme Activation , Female , Humans , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/pathology , Lung/blood supply , Lung/pathology , Macrophages, Alveolar/metabolism , Male , Middle Aged , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/pathology , Recombinant Proteins/metabolism , Scleroderma, Systemic/complications , Scleroderma, Systemic/pathology , T-Lymphocytes/metabolism , Up-Regulation
20.
Am J Physiol Lung Cell Mol Physiol ; 296(4): L582-93, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19136574

ABSTRACT

Pulmonary hypertension (PH) is a serious disease of multiple etiologies mediated by hypoxia, immune stimuli, and elevated pulmonary pressure that leads to vascular thickening and eventual right heart failure. In a chronic hypoxia model of PH, we previously reported the induction of a novel pleiotropic cytokine, hypoxia-induced mitogenic factor (HIMF), that exhibits mitogenic, vasculogenic, contractile, and chemokine properties during PH-associated vascular remodeling. To examine the role of HIMF in hypoxia-induced vascular remodeling, we performed in vivo knockdown of HIMF using short hairpin RNA directed at rat HIMF in the chronic hypoxia model of PH. Knockdown of HIMF partially blocked increases in mean pulmonary artery pressure, pulmonary vascular resistance, right heart hypertrophy, and vascular remodeling caused by chronic hypoxia. To demonstrate a direct role for HIMF in the mechanism of PH development, we performed HIMF-gene transfer into the lungs of rats using a HIMF-expressing adeno-associated virus (AAV). AAV-HIMF alone caused development of PH similar to that of chronic hypoxia with increased mean pulmonary artery pressure and pulmonary vascular resistance, right heart hypertrophy, and neomuscularization and thickening of small pulmonary arterioles. The findings suggest that HIMF represents a critical cytokine-like growth factor in the development of PH.


Subject(s)
Blood Vessels/physiopathology , Hemodynamics/physiology , Hypertension, Pulmonary/complications , Hypertension, Pulmonary/physiopathology , Hypoxia/complications , Nerve Growth Factor/metabolism , Animals , Cell Line , Chronic Disease , Disease Models, Animal , Gene Knockdown Techniques , Gene Transfer Techniques , Humans , Hypertension, Pulmonary/pathology , Lung/pathology , Lung/physiopathology , Male , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...