Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Lymphat Res Biol ; 22(3): 195-202, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38699876

ABSTRACT

Background: Lymphedema is chronic limb swelling resulting from lymphatic dysfunction. It affects an estimated five million Americans. There is no cure for this disease. Assessing lymphatic growth is essential in developing novel therapeutics. Intravital microscopy (IVM) is a powerful imaging tool for investigating various biological processes in live animals. Tissue nanotransfection technology (TNT) facilitates a direct, transcutaneous nonviral vector gene delivery using a chip with nanochannel poration in a rapid (<100 ms) focused electric field. TNT was used in this study to deliver the genetic cargo in the murine tail lymphedema to assess the lymphangiogenesis. The purpose of this study is to experimentally evaluate the applicability of IVM to visualize and quantify lymphatics in the live mice model. Methods and Results: The murine tail model of lymphedema was utilized. TNT was applied to the murine tail (day 0) directly at the surgical site with genetic cargo loaded into the TNT reservoir: TNTpCMV6 group receives pCMV6 (expression vector backbone alone) (n = 6); TNTProx1 group receives pCMV6-Prox1 (n = 6). Lymphatic vessels (fluorescein isothiocyanate [FITC]-dextran stained) and lymphatic branch points (indicating lymphangiogenesis) were analyzed with the confocal/multiphoton microscope. The experimental group TNTProx1 exhibited reduced postsurgical tail lymphedema and increased lymphatic distribution compared to TNTpCMV6 group. More lymphatic branching points (>3-fold) were observed at the TNT site in TNTProx1 group. Conclusions: This study demonstrates a novel, powerful imaging tool for investigating lymphatic vessels in live murine tail model of lymphedema. IVM can be utilized for functional assessment of lymphatics and visualization of lymphangiogenesis following gene-based therapy.


Subject(s)
Disease Models, Animal , Intravital Microscopy , Lymphangiogenesis , Lymphatic Vessels , Lymphedema , Tail , Animals , Lymphedema/pathology , Lymphedema/diagnostic imaging , Lymphedema/metabolism , Lymphedema/genetics , Mice , Intravital Microscopy/methods , Lymphatic Vessels/diagnostic imaging , Lymphatic Vessels/pathology , Lymphatic Vessels/metabolism , Female , Gene Transfer Techniques
2.
Blood ; 142(3): 290-305, 2023 07 20.
Article in English | MEDLINE | ID: mdl-37192286

ABSTRACT

Despite >80 years of clinical experience with coagulation factor VIII (FVIII) inhibitors, surprisingly little is known about the in vivo mechanism of this most serious complication of replacement therapy for hemophilia A. These neutralizing antidrug alloantibodies arise in ∼30% of patients. Inhibitor formation is T-cell dependent, but events leading up to helper T-cell activation have been elusive because of, in part, the complex anatomy and cellular makeup of the spleen. Here, we show that FVIII antigen presentation to CD4+ T cells critically depends on a select set of several anatomically distinct antigen-presenting cells, whereby marginal zone B cells and marginal zone and marginal metallophilic macrophages but not red pulp macrophages (RPMFs) participate in shuttling FVIII to the white pulp in which conventional dendritic cells (DCs) prime helper T cells, which then differentiate into follicular helper T (Tfh) cells. Toll-like receptor 9 stimulation accelerated Tfh cell responses and germinal center and inhibitor formation, whereas systemic administration of FVIII alone in hemophilia A mice increased frequencies of monocyte-derived and plasmacytoid DCs. Moreover, FVIII enhanced T-cell proliferation to another protein antigen (ovalbumin), and inflammatory signaling-deficient mice were less likely to develop inhibitors, indicating that FVIII may have intrinsic immunostimulatory properties. Ovalbumin, which, unlike FVIII, is absorbed into the RPMF compartment, fails to elicit T-cell proliferative and antibody responses when administered at the same dose as FVIII. Altogether, we propose that an antigen trafficking pattern that results in efficient in vivo delivery to DCs and inflammatory signaling, shape the immunogenicity of FVIII.


Subject(s)
CD4-Positive T-Lymphocytes , Factor VIII , Hemophilia A , Hemostatics , Animals , Mice , Dendritic Cells/metabolism , Factor VIII/immunology , Factor VIII/therapeutic use , Hemophilia A/drug therapy , Hemostatics/immunology , Hemostatics/therapeutic use , Ovalbumin/immunology
3.
Front Endocrinol (Lausanne) ; 13: 989844, 2022.
Article in English | MEDLINE | ID: mdl-36568089

ABSTRACT

Type 2 diabetes mellitus (T2DM) causes peripheral vascular disease because of which several blood-borne factors, including vital nutrients fail to reach the affected tissue. Tissue epigenome is sensitive to chronic hyperglycemia and is known to cause pathogenesis of micro- and macrovascular complications. These vascular complications of T2DM may perpetuate the onset of organ dysfunction. The burden of diabetes is primarily because of a wide range of complications of which nonhealing diabetic ulcers represent a major component. Thus, it is imperative that current research help recognize more effective methods for the diagnosis and management of early vascular injuries. This review addresses the significance of epigenetic processes such as DNA methylation and histone modifications in the evolution of macrovascular and microvascular complications of T2DM.


Subject(s)
Diabetes Mellitus, Type 2 , Diabetic Angiopathies , Vascular Diseases , Humans , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Diabetic Angiopathies/genetics , Diabetic Angiopathies/complications , Epigenesis, Genetic , DNA Methylation , Vascular Diseases/complications
4.
J Cell Biochem ; 123(11): 1817-1826, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35959632

ABSTRACT

Understanding the mechanisms responsible for the kidney's defense against ascending uropathogen is critical to devise novel treatment strategies against increasingly antibiotic resistant uropathogen. Growing body of evidence indicate Intercalated cells of the kidney as the key innate immune epithelial cells against uropathogen. The aim of this study was to find orthologous and differentially expressed bacterial defense genes in human versus murine intercalated cells. We simultaneously analyzed 84 antibacterial genes in intercalated cells enriched from mouse and human kidney samples. Intercalated cell "reporter mice" were exposed to saline versus uropathogenic Escherichia coli (UPEC) transurethrally for 1 h in vivo, and intercalated cells were flow sorted. Human kidney intercalated cells were enriched from kidney biopsy using magnetic-activated cell sorting and exposed to UPEC in vitro for 1 h. RT2 antibacterial PCR array was performed. Mitogen-activated protein kinase kinase kinase 7 (MAP3K7) messenger RNA (mRNA) expression increased in intercalated cells of both humans and mice following UPEC exposure. Intercalated cell MAP3K7 protein expression was defined by immunofluorescence and confocal imaging analysis, was consistent with the increased MAP3K7 mRNA expression profiles defined by PCR. The presence of the orthologous innate immune gene MAP3K7/TAK1 suggests that it may be a key regulator of the intercalated cell antibacterial response and demands further investigation of its role in urinary tract infection pathogenesis.


Subject(s)
Escherichia coli Infections , Uropathogenic Escherichia coli , Humans , Mice , Animals , Uropathogenic Escherichia coli/genetics , Escherichia coli Infections/genetics , Escherichia coli Infections/microbiology , Kidney , Epithelial Cells/microbiology , Genes, Regulator , Immunity, Innate/genetics , Anti-Bacterial Agents , RNA, Messenger
5.
Am J Physiol Renal Physiol ; 321(6): F675-F688, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34658261

ABSTRACT

Expansion of renal lymphatic networks, or lymphangiogenesis (LA), is well recognized during development and is now being implicated in kidney diseases. Although LA is associated with multiple pathological conditions, very little is known about its role in acute kidney injury. The purpose of this study was to evaluate the role of LA in a model of cisplatin-induced nephrotoxicity. LA is predominately regulated by vascular endothelial growth factor (VEGF)-C and VEGF-D, ligands that exert their function through their cognate receptor VEGF receptor 3 (VEGFR3). We demonstrated that use of MAZ51, a selective VEGFR3 inhibitor, caused significantly worse structural and functional kidney damage in cisplatin nephrotoxicity. Apoptotic cell death and inflammation were also increased in MAZ51-treated animals compared with vehicle-treated animals following cisplatin administration. Notably, MAZ51 caused significant upregulation of intrarenal phospho-NF-κB, phospho-JNK, and IL-6. Cisplatin nephrotoxicity is associated with vascular congestion due to endothelial dysfunction. Using three-dimensional tissue cytometry, a novel approach to explore lymphatics in the kidney, we detected significant vascular autofluorescence attributed to erythrocytes in cisplatin alone-treated animals. Interestingly, no such congestion was detected in MAZ51-treated animals. We found increased renal vascular damage in MAZ51-treated animals, whereby MAZ51 caused a modest decrease in the endothelial markers endomucin and von Willebrand factor, with a modest increase in VEGFR2. Our findings identify a protective role for de novo LA in cisplatin nephrotoxicity and provide a rationale for the development of therapeutic approaches targeting LA. Our study also suggests off-target effects of MAZ51 on the vasculature in the setting of cisplatin nephrotoxicity.NEW & NOTEWORTHY Little is known about injury-associated LA in the kidney and its role in the pathophysiology of acute kidney injury (AKI). Observed exacerbation of cisplatin-induced AKI after LA inhibition was accompanied by increased medullary damage and cell death in the kidney. LA inhibition also upregulated compensatory expression of LA regulatory proteins, including JNK and NF-κB. These data support the premise that LA is induced during AKI and lymphatic expansion is a protective mechanism in cisplatin nephrotoxicity.


Subject(s)
Indoles/toxicity , Kidney Diseases/chemically induced , Kidney/drug effects , Lymphangiogenesis/drug effects , Lymphatic Vessels/drug effects , Naphthalenes/toxicity , Protein Kinase Inhibitors/toxicity , Vascular Endothelial Growth Factor Receptor-3/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cisplatin , Disease Models, Animal , Inflammation Mediators/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Kidney/enzymology , Kidney/pathology , Kidney/physiopathology , Kidney Diseases/enzymology , Kidney Diseases/pathology , Kidney Diseases/physiopathology , Lymphatic Vessels/enzymology , Lymphatic Vessels/pathology , Lymphatic Vessels/physiopathology , Male , Mice, Inbred C57BL , NF-kappa B/metabolism , Phosphorylation , Signal Transduction , Vascular Endothelial Growth Factor Receptor-3/metabolism
6.
Lab Invest ; 101(9): 1186-1196, 2021 09.
Article in English | MEDLINE | ID: mdl-34017058

ABSTRACT

The lymphatic system plays an integral role in physiology and has recently been identified as a key player in disease progression. Tissue injury stimulates lymphatic expansion, or lymphangiogenesis (LA), though its precise role in disease processes remains unclear. LA is associated with inflammation, which is a key component of acute kidney injury (AKI), for which there are no approved therapies. While LA research has gained traction in the last decade, there exists a significant lack of understanding of this process in the kidney. Though innovative studies have elucidated markers and models with which to study LA, the field is still evolving with ways to visualize lymphatics in vivo. Prospero-related homeobox-1 (Prox-1) is the master regulator of LA and determines lymphatic cell fate through its action on vascular endothelial growth factor receptor expression. Here, we investigate the consequences of AKI on the abundance and distribution of lymphatic endothelial cells using Prox1-tdTomato reporter mice (ProxTom) coupled with large-scale three-dimensional quantitative imaging and tissue cytometry (3DTC). Using these technologies, we describe the spatial dynamics of lymphatic vasculature in quiescence and post-AKI. We also describe the use of lymphatic vessel endothelial hyaluronan receptor-1 (LYVE-1) as a marker of lymphatic vessels using 3DTC in the absence of the ProxTom reporter mice as an alternative approach. The use of 3DTC for lymphatic research presents a new avenue with which to study the origin and distribution of renal lymphatic vessels. These findings will enhance our understanding of renal lymphatic function during injury and could inform the development of novel therapeutics for intervention in AKI.


Subject(s)
Acute Kidney Injury , Image Cytometry , Imaging, Three-Dimensional , Lymphatic Vessels , Acute Kidney Injury/diagnostic imaging , Acute Kidney Injury/metabolism , Animals , Homeodomain Proteins/metabolism , Lymphangiogenesis , Lymphatic Vessels/diagnostic imaging , Lymphatic Vessels/metabolism , Male , Membrane Transport Proteins/metabolism , Mice , Mice, Transgenic , Tumor Suppressor Proteins/metabolism
7.
Nat Commun ; 12(1): 2405, 2021 04 23.
Article in English | MEDLINE | ID: mdl-33893305

ABSTRACT

Kidney intercalated cells are involved in acid-base homeostasis via vacuolar ATPase expression. Here we report six human intercalated cell subtypes, including hybrid principal-intercalated cells identified from single cell transcriptomics. Phagosome maturation is a biological process that increases in biological pathway analysis rank following exposure to uropathogenic Escherichia coli in two of the intercalated cell subtypes. Real time confocal microscopy visualization of murine renal tubules perfused with green fluorescent protein expressing Escherichia coli or pHrodo Green E. coli BioParticles demonstrates that intercalated cells actively phagocytose bacteria then acidify phagolysosomes. Additionally, intercalated cells have increased vacuolar ATPase expression following in vivo experimental UTI. Taken together, intercalated cells exhibit a transcriptional response conducive to the kidney's defense, engulf bacteria and acidify the internalized bacteria. Intercalated cells represent an epithelial cell with characteristics of professional phagocytes like macrophages.


Subject(s)
Epithelial Cells/immunology , Kidney/immunology , Phagocytes/immunology , Uropathogenic Escherichia coli/immunology , Acids/chemistry , Animals , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Profiling/methods , Humans , Kidney/cytology , Kidney/metabolism , Kidney Tubules, Collecting/cytology , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Phagocytes/cytology , Single-Cell Analysis/methods , Vacuolar Proton-Translocating ATPases/metabolism
8.
Lab Invest ; 101(5): 661-676, 2021 05.
Article in English | MEDLINE | ID: mdl-33408350

ABSTRACT

The advent of personalized medicine has driven the development of novel approaches for obtaining detailed cellular and molecular information from clinical tissue samples. Tissue cytometry is a promising new technique that can be used to enumerate and characterize each cell in a tissue and, unlike flow cytometry and other single-cell techniques, does so in the context of the intact tissue, preserving spatial information that is frequently crucial to understanding a cell's physiology, function, and behavior. However, the wide-scale adoption of tissue cytometry as a research tool has been limited by the fact that published examples utilize specialized techniques that are beyond the capabilities of most laboratories. Here we describe a complete and accessible pipeline, including methods of sample preparation, microscopy, image analysis, and data analysis for large-scale three-dimensional tissue cytometry of human kidney tissues. In this workflow, multiphoton microscopy of unlabeled tissue is first conducted to collect autofluorescence and second-harmonic images. The tissue is then labeled with eight fluorescent probes, and imaged using spectral confocal microscopy. The raw 16-channel images are spectrally deconvolved into 8-channel images, and analyzed using the Volumetric Tissue Exploration and Analysis (VTEA) software developed by our group. We applied this workflow to analyze millimeter-scale tissue samples obtained from human nephrectomies and from renal biopsies from individuals diagnosed with diabetic nephropathy, generating a quantitative census of tens of thousands of cells in each. Such analyses can provide useful insights that can be linked to the biology or pathology of kidney disease. The approach utilizes common laboratory techniques, is compatible with most commercially-available confocal microscope systems and all image and data analysis is conducted using the VTEA image analysis software, which is available as a plug-in for ImageJ.


Subject(s)
Cytological Techniques , Imaging, Three-Dimensional , Kidney/cytology , Microscopy, Fluorescence, Multiphoton , Software , Fluorescent Dyes , Humans , Microscopy, Confocal
9.
JCI Insight ; 5(9)2020 05 07.
Article in English | MEDLINE | ID: mdl-32376804

ABSTRACT

Immune checkpoint blockade immunotherapy delivers promising clinical results in colorectal cancer (CRC). However, only a fraction of cancer patients develop durable responses. The tumor microenvironment (TME) negatively impacts tumor immunity and subsequently clinical outcomes. Therefore, there is a need to identify other checkpoint targets associated with the TME. Early-onset factors secreted by stromal cells as well as tumor cells often help recruit immune cells to the TME, among which are alarmins such as IL-33. The only known receptor for IL-33 is stimulation 2 (ST2). Here we demonstrated that high ST2 expression is associated with poor survival and is correlated with low CD8+ T cell cytotoxicity in CRC patients. ST2 is particularly expressed in tumor-associated macrophages (TAMs). In preclinical models of CRC, we demonstrated that ST2-expressing TAMs (ST2+ TAMs) were recruited into the tumor via CXCR3 expression and exacerbated the immunosuppressive TME; and that combination of ST2 depletion using ST2-KO mice with anti-programmed death 1 treatment resulted in profound growth inhibition of CRC. Finally, using the IL-33trap fusion protein, we suppressed CRC tumor growth and decreased tumor-infiltrating ST2+ TAMs. Together, our findings suggest that ST2 could serve as a potential checkpoint target for CRC immunotherapy.


Subject(s)
Colorectal Neoplasms/immunology , Interleukin-1 Receptor-Like 1 Protein/immunology , Interleukin-33/immunology , Tumor Microenvironment/immunology , Tumor-Associated Macrophages/immunology , Animals , Cell Line, Tumor , Databases, Genetic , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Tumor-Associated Macrophages/cytology
10.
Sci Rep ; 9(1): 8449, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31186447

ABSTRACT

The pancreatic islet is a complex micro-organ containing numerous cell types, including endocrine, immune, and endothelial cells. The communication of these systems is lost upon isolation of the islets, and therefore the pathogenesis of diabetes can only be fully understood by studying this organized, multicellular environment in vivo. We have developed several adaptable tools to create a versatile platform to interrogate ß-cell function in vivo. Specifically, we developed ß-cell-selective virally-encoded fluorescent protein biosensors that can be rapidly and easily introduced into any mouse. We then coupled the use of these biosensors with intravital microscopy, a powerful tool that can be used to collect cellular and subcellular data from living tissues. Together, these approaches allowed the observation of in vivo ß-cell-specific ROS dynamics using the Grx1-roGFP2 biosensor and calcium signaling using the GcAMP6s biosensor. Next, we utilized abdominal imaging windows (AIW) to extend our in vivo observations beyond single-point terminal measurements to collect longitudinal physiological and biosensor data through repeated imaging of the same mice over time. This platform represents a significant advancement in our ability to study ß-cell structure and signaling in vivo, and its portability for use in virtually any mouse model will enable meaningful studies of ß-cell physiology in the endogenous islet niche.


Subject(s)
Endothelial Cells/ultrastructure , Insulin-Secreting Cells/ultrastructure , Intravital Microscopy/methods , Islets of Langerhans/ultrastructure , Animals , Biosensing Techniques , Calcium Signaling/genetics , Calcium Signaling/immunology , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Islets of Langerhans Transplantation , Mice
11.
Sci Rep ; 8(1): 13759, 2018 09 13.
Article in English | MEDLINE | ID: mdl-30214007

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) has reactive stroma that promotes tumor signaling, fibrosis, inflammation, and hypoxia, which activates HIF-1α to increase tumor cell metastasis and therapeutic resistance. Carbonic anhydrase IX (CA9) stabilizes intracellular pH following induction by HIF-1α. Redox effector factor-1 (APE1/Ref-1) is a multifunctional protein with redox signaling activity that converts certain oxidized transcription factors to a reduced state, enabling them to upregulate tumor-promoting genes. Our studies evaluate PDAC hypoxia responses and APE1/Ref-1 redox signaling contributions to HIF-1α-mediated CA9 transcription. Our previous studies implicated this pathway in PDAC cell survival under hypoxia. We expand those studies, comparing drug responses using patient-derived PDAC cells displaying differential hypoxic responses in 3D spheroid tumor-stroma models to characterize second generation APE1/Ref-1 redox signaling and CA9 inhibitors. Our data demonstrates that HIF-1α-mediated CA9 induction differs between patient-derived PDAC cells and that APE1/Ref-1 redox inhibition attenuates this induction by decreasing hypoxia-induced HIF-1 DNA binding. Dual-targeting of APE1/Ref-1 and CA9 in 3D spheroids demonstrated that this combination effectively kills PDAC tumor cells displaying drastically different levels of CA9. New APE1/Ref-1 and CA9 inhibitors were significantly more potent alone and in combination, highlighting the potential of combination therapy targeting the APE1-Ref-1 signaling axis with significant clinical potential.


Subject(s)
Antigens, Neoplasm/genetics , Carbonic Anhydrase IX/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Pancreatic Neoplasms/drug therapy , Carbonic Anhydrase IX/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival , DNA-(Apurinic or Apyrimidinic Site) Lyase/antagonists & inhibitors , Gene Expression Regulation, Neoplastic/genetics , Humans , Pancreatic Neoplasms/genetics , Signal Transduction/drug effects , Small Molecule Libraries/pharmacology , Transcription, Genetic/drug effects
12.
J Clin Invest ; 127(12): 4527-4540, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29130940

ABSTRACT

Endothelial cells (ECs) are components of the hematopoietic microenvironment and regulate hematopoietic stem and progenitor cell (HSPC) homeostasis. Cytokine treatments that cause HSPC trafficking to peripheral blood are associated with an increase in dipeptidylpeptidase 4/CD26 (DPP4/CD26), an enzyme that truncates the neurotransmitter neuropeptide Y (NPY). Here, we show that enzymatically altered NPY signaling in ECs caused reduced VE-cadherin and CD31 expression along EC junctions, resulting in increased vascular permeability and HSPC egress. Moreover, selective NPY2 and NPY5 receptor antagonists restored vascular integrity and limited HSPC mobilization, demonstrating that the enzymatically controlled vascular gateway specifically opens by cleavage of NPY by CD26 signaling via NPY2 and NPY5 receptors. Mice lacking CD26 or NPY exhibited impaired HSPC trafficking that was restored by treatment with truncated NPY. Thus, our results point to ECs as gatekeepers of HSPC trafficking and identify a CD26-mediated NPY axis that has potential as a pharmacologic target to regulate hematopoietic trafficking in homeostatic and stress conditions.


Subject(s)
Dipeptidyl Peptidase 4/metabolism , Endothelial Cells/metabolism , Hematopoietic Stem Cells/metabolism , Neuropeptide Y/metabolism , Signal Transduction/physiology , Animals , Dipeptidyl Peptidase 4/genetics , Endothelial Cells/cytology , Hematopoietic Stem Cells/cytology , Humans , Mice , Mice, Knockout , Neuropeptide Y/genetics , Receptors, Neuropeptide Y/agonists , Receptors, Neuropeptide Y/genetics , Receptors, Neuropeptide Y/metabolism
13.
J Vis Exp ; (121)2017 03 21.
Article in English | MEDLINE | ID: mdl-28362378

ABSTRACT

Increasing evidence indicates that normal hematopoiesis is regulated by distinct microenvironmental cues in the BM, which include specialized cellular niches modulating critical hematopoietic stem cell (HSC) functions1,2. Indeed, a more detailed picture of the hematopoietic microenvironment is now emerging, in which the endosteal and the endothelial niches form functional units for the regulation of normal HSC and their progeny3,4,5. New studies have revealed the importance of perivascular cells, adipocytes and neuronal cells in maintaining and regulating HSC function6,7,8. Furthermore, there is evidence that cells from different lineages, i.e. myeloid and lymphoid cells, home and reside in specific niches within the BM microenvironment. However, a complete mapping of the BM microenvironment and its occupants is still in progress. Transgenic mouse strains expressing lineage specific fluorescent markers or mice genetically engineered to lack selected molecules in specific cells of the BM niche are now available. Knock-out and lineage tracking models, in combination with transplantation approaches, provide the opportunity to refine the knowledge on the role of specific "niche" cells for defined hematopoietic populations, such as HSC, B-cells, T-cells, myeloid cells and erythroid cells. This strategy can be further potentiated by merging the use of two-photon microscopy of the calvarium. By providing in vivo high resolution imaging and 3-D rendering of the BM calvarium, we can now determine precisely the location where specific hematopoietic subsets home in the BM and evaluate the kinetics of their expansion over time. Here, Lys-GFP transgenic mice (marking myeloid cells)9 and RBPJ knock-out mice (lacking canonical Notch signaling)10 are used in combination with IVFM to determine the engraftment of myeloid cells to a Notch defective BM microenvironment.


Subject(s)
Bone Marrow Cells/cytology , Hematopoietic Stem Cells/cytology , Intravital Microscopy/methods , Microscopy, Fluorescence/methods , Models, Genetic , Stem Cell Niche , Animals , Bone Marrow Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction
14.
Am J Physiol Renal Physiol ; 313(2): F163-F173, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28404591

ABSTRACT

Hypertension is one of the most prevalent diseases worldwide and a major risk factor for renal failure and cardiovascular disease. The role of albuminuria, a common feature of hypertension and robust predictor of cardiorenal disorders, remains incompletely understood. The goal of this study was to investigate the mechanisms leading to albuminuria in the kidney of a rat model of hypertension, the Dahl salt-sensitive (SS) rat. To determine the relative contributions of the glomerulus and proximal tubule (PT) to albuminuria, we applied intravital two-photon-based imaging to investigate the complex renal physiological changes that occur during salt-induced hypertension. Following a high-salt diet, SS rats exhibited elevated blood pressure, increased glomerular sieving of albumin (GSCalb = 0.0686), relative permeability to albumin (+Δ16%), and impaired volume hemodynamics (-Δ14%). Serum albumin but not serum globulins or creatinine concentration was decreased (-0.54 g/dl), which was concomitant with increased filtration of albumin (3.7 vs. 0.8 g/day normal diet). Pathologically, hypertensive animals had significant tubular damage, as indicated by increased prevalence of granular casts, expansion and necrosis of PT epithelial cells (+Δ2.20 score/image), progressive augmentation of red blood cell velocity (+Δ269 µm/s) and micro vessel diameter (+Δ4.3 µm), and increased vascular injury (+Δ0.61 leakage/image). Therefore, development of salt-induced hypertension can be triggered by fast and progressive pathogenic remodeling of PT epithelia, which can be associated with changes in albumin handling. Collectively, these results indicate that both the glomerulus and the PT contribute to albuminuria, and dual treatment of glomerular filtration and albumin reabsorption may represent an effective treatment of salt-sensitive hypertension.


Subject(s)
Albuminuria/etiology , Blood Pressure , Hypertension/etiology , Intravital Microscopy , Kidney Glomerulus/pathology , Kidney Tubules, Proximal/pathology , Microscopy, Fluorescence, Multiphoton , Albuminuria/blood , Albuminuria/pathology , Albuminuria/physiopathology , Animals , Disease Models, Animal , Glomerular Filtration Rate , Hypertension/blood , Hypertension/pathology , Hypertension/physiopathology , Kidney Glomerulus/metabolism , Kidney Glomerulus/physiopathology , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/physiopathology , Rats, Inbred Dahl , Renal Reabsorption , Serum Albumin/metabolism , Sodium Chloride, Dietary , Time Factors
15.
J Neurosurg ; 127(6): 1219-1230, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28059653

ABSTRACT

OBJECTIVE Defects in the apoptotic machinery and augmented survival signals contribute to drug resistance in glioblastoma (GBM). Moreover, another complexity related to GBM treatment is the concept that GBM development and recurrence may arise from the expression of GBM stem cells (GSCs). Therefore, the use of a multifaceted approach or multitargeted agents that affect specific tumor cell characteristics will likely be necessary to successfully eradicate GBM. The objective of this study was to investigate the usefulness of sulforaphane (SFN)-a constituent of cruciferous vegetables with a multitargeted effect-as a therapeutic agent for GBM. METHODS The inhibitory effects of SFN on established cell lines, early primary cultures, CD133-positive GSCs, GSC-derived spheroids, and GBM xenografts were evaluated using various methods, including GSC isolation and the sphere-forming assay, analysis of reactive oxygen species (ROS) and apoptosis, cell growth inhibition assay, comet assays for assessing SFN-triggered DNA damage, confocal microscopy, Western blot analysis, and the determination of in vivo efficacy as assessed in human GBM xenograft models. RESULTS SFN triggered the significant inhibition of cell survival and induced apoptotic cell death, which was associated with caspase 3 and caspase 7 activation. Moreover, SFN triggered the formation of mitochondrial ROS, and SFN-triggered cell death was ROS dependent. Comet assays revealed that SFN increased single- and double-strand DNA breaks in GBM. Compared with the vehicle control cells, a significantly higher amount of γ-H2AX foci correlated with an increase in DNA double-strand breaks in the SFN-treated samples. Furthermore, SFN robustly inhibited the growth of GBM cell-induced cell death in established cell cultures and early-passage primary cultures and, most importantly, was effective in eliminating GSCs, which play a major role in drug resistance and disease recurrence. In vivo studies revealed that SFN administration at 100 mg/kg for 5-day cycles repeated for 3 weeks significantly decreased the growth of ectopic xenografts that were established from the early passage of primary cultures of GBM10. CONCLUSIONS These results suggest that SFN is a potent anti-GBM agent that targets several apoptosis and cell survival pathways and further preclinical and clinical studies may prove that SFN alone or in combination with other therapies may be potentially useful for GBM therapy.


Subject(s)
Anticarcinogenic Agents/pharmacology , Cell Survival/drug effects , Glioblastoma/metabolism , Isothiocyanates/pharmacology , Neoplastic Stem Cells/drug effects , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Cell Line, Tumor , DNA Damage/drug effects , Glioblastoma/pathology , Humans , Mice , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Sulfoxides
16.
J Endod ; 42(10): 1490-5, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27663615

ABSTRACT

INTRODUCTION: Root canal disinfection and the establishment of an intracanal microenvironment conducive to the proliferation/differentiation of stem cells play a significant role in regenerative endodontics. This study was designed to (1) investigate the antimicrobial efficacy of triple antibiotic-containing nanofibers against a dual-species biofilm and (2) evaluate the ability of dental pulp stem cells (DPSCs) to adhere to and proliferate on dentin upon nanofiber exposure. METHODS: Seven-day-old dual-species biofilm established on dentin specimens was exposed for 3 days to the following: saline (control), antibiotic-free nanofibers (control), and triple antibiotic-containing nanofibers or a saturated triple antibiotic paste (TAP) solution (50 mg/mL in phosphate buffer solution). Bacterial viability was assessed using the LIVE/DEAD assay (Molecular Probes, Inc, Eugene, OR) and confocal laser scanning microscopy. For cytocompatibility studies, dentin specimens after nanofiber or TAP (1 g/mL in phosphate buffer solution) exposure were evaluated for cell adhesion and spreading by actin-phalloidin staining. DPSC proliferation was assessed on days 1, 3, and 7. Statistics were performed, and significance was set at the 5% level. RESULTS: Confocal laser scanning microscopy showed significant bacterial death upon antibiotic-containing nanofiber exposure, differing significantly (P < .05) from antibiotic-free fibers and the control (saline). DPSCs showed enhanced adhesion/spreading on dentin specimens treated with antibiotic-containing nanofibers when compared with its TAP counterparts. The DPSC proliferation rate was similar on days 1 and 3 in antibiotic-free nanofibers, triple antibiotic-containing nanofibers, and TAP-treated dentin. Proliferation was higher (9-fold) on dentin treated with antibiotic-containing nanofibers on day 7 compared with TAP. CONCLUSIONS: Triple antibiotic-containing polymer nanofibers led to significant bacterial death, whereas they did not affect DPSC attachment and proliferation on dentin.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Biofilms/drug effects , Drug Delivery Systems/methods , Nanofibers/administration & dosage , Polymers/administration & dosage , Anti-Bacterial Agents/chemistry , Bacteria/cytology , Bacteria/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Dental Pulp/cytology , Dental Pulp/drug effects , Dental Pulp/microbiology , Dentin/microbiology , Disinfection/methods , Humans , Nanofibers/chemistry , Root Canal Therapy/methods , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/microbiology
17.
Mol Cancer Ther ; 15(5): 794-805, 2016 05.
Article in English | MEDLINE | ID: mdl-26873728

ABSTRACT

Constitutively activated STAT3 protein has been found to be a key regulator of pancreatic cancer and a target for molecular therapeutic intervention. In this study, PG-S3-001, a small molecule derived from the SH-4-54 class of STAT3 inhibitors, was found to inhibit patient-derived pancreatic cancer cell proliferation in vitro and in vivo in the low micromolar range. PG-S3-001 binds the STAT3 protein potently, Kd = 324 nmol/L by surface plasmon resonance, and showed no effect in a kinome screen (>100 cancer-relevant kinases). In vitro studies demonstrated potent cell killing as well as inhibition of STAT3 activation in pancreatic cancer cells. To better model the tumor and its microenvironment, we utilized three-dimensional (3D) cultures of patient-derived pancreatic cancer cells in the absence and presence of cancer-associated fibroblasts (CAF). In this coculture model, inhibition of tumor growth is maintained following STAT3 inhibition in the presence of CAFs. Confocal microscopy was used to verify tumor cell death following treatment of 3D cocultures with PG-S3-001. The 3D model was predictive of in vivo efficacy as significant tumor growth inhibition was observed upon administration of PG-S3-001. These studies showed that the inhibition of STAT3 was able to impact the survival of tumor cells in a relevant 3D model, as well as in a xenograft model using patient-derived cells. Mol Cancer Ther; 15(5); 794-805. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Female , Humans , Ligands , Male , Models, Molecular , Molecular Conformation , Pancreatic Neoplasms/drug therapy , Phosphorylation , Protein Binding , STAT3 Transcription Factor/chemistry , Signal Transduction/drug effects , Structure-Activity Relationship , Xenograft Model Antitumor Assays , src Homology Domains
18.
PLoS One ; 10(7): e0131677, 2015.
Article in English | MEDLINE | ID: mdl-26136112

ABSTRACT

Autosomal recessive polycystic kidney disease is a truly catastrophic monogenetic disease, causing death and end stage renal disease in neonates and children. Using PCK female rats, an orthologous model of autosomal recessive polycystic kidney disease harboring mutant Pkhd1, we tested the hypothesis that intravenous renal cell transplantation with normal Sprague Dawley male kidney cells would improve the polycystic kidney disease phenotype. Cytotherapy with renal cells expressing wild type Pkhd1 and tubulogenic serum amyloid A1 had powerful and sustained beneficial effects on renal function and structure in the polycystic kidney disease model. Donor cell engraftment and both mutant and wild type Pkhd1 were found in treated but not control PCK kidneys 15 weeks after the final cell infusion. To examine the mechanisms of global protection with a small number of transplanted cells, we tested the hypothesis that exosomes derived from normal Sprague Dawley cells can limit the cystic phenotype of PCK recipient cells. We found that renal exosomes originating from normal Sprague Dawley cells carried and transferred wild type Pkhd1 mRNA to PCK cells in vivo and in vitro and restricted cyst formation by cultured PCK cells. The results indicate that transplantation with renal cells containing wild type Pkhd1 improves renal structure and function in autosomal recessive polycystic kidney disease and may provide an intra-renal supply of normal Pkhd1 mRNA.


Subject(s)
Kidney Tubules/pathology , Polycystic Kidney, Autosomal Recessive/metabolism , Polycystic Kidney, Autosomal Recessive/therapy , Animals , Cell Proliferation/drug effects , Cell- and Tissue-Based Therapy , Cells, Cultured , Cysts/metabolism , Disease Models, Animal , Exosomes , Female , Genotype , Immunohistochemistry , In Situ Hybridization, Fluorescence , Liver/metabolism , Male , Phenotype , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Serum Amyloid A Protein/metabolism
19.
Am J Physiol Lung Cell Mol Physiol ; 309(4): L425-34, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26092999

ABSTRACT

In addition to exerting a potent anti-elastase function, α-1 antitrypsin (A1AT) maintains the structural integrity of the lung by inhibiting endothelial inflammation and apoptosis. A main serpin secreted in circulation by hepatocytes, A1AT requires uptake by the endothelium to achieve vasculoprotective effects. This active uptake mechanism, which is inhibited by cigarette smoking (CS), involves primarily clathrin- but also caveola-mediated endocytosis and may require active binding to a receptor. Because circulating A1AT binds to high-density lipoprotein (HDL), we hypothesized that scavenging receptors are candidates for endothelial uptake of the serpin. Although the low-density lipoprotein (LDL) receptor-related protein 1 (LRP1) internalizes only elastase-bound A1AT, the scavenger receptor B type I (SR-BI), which binds and internalizes HDL and is modulated by CS, may be involved in A1AT uptake. Transmission electron microscopy imaging of colloidal gold-labeled A1AT confirmed A1AT endocytosis in both clathrin-coated vesicles and caveolae in endothelial cells. SR-BI immunoprecipitation identified binding to A1AT at the plasma membrane. Pretreatment of human lung microvascular endothelial cells with SR-B ligands (HDL or LDL), knockdown of SCARB1 expression, or neutralizing SR-BI antibodies significantly reduced A1AT uptake by 30-50%. Scarb1 null mice exhibited decreased A1AT lung content following systemic A1AT administration and reduced lung anti-inflammatory effects of A1AT supplementation during short-term CS exposure. In turn, A1AT supplementation increased lung SR-BI expression and modulated circulating lipoprotein levels in wild-type animals. These studies indicate that SR-BI is an important mediator of A1AT endocytosis in pulmonary endothelium and suggest a cross talk between A1AT and lipoprotein regulation of vascular functions.


Subject(s)
Endothelial Cells/metabolism , Scavenger Receptors, Class B/physiology , Smoking/metabolism , alpha 1-Antitrypsin/metabolism , Animals , Binding, Competitive , Cells, Cultured , Endocytosis , Endothelium, Vascular/pathology , Female , Humans , Male , Mice, Inbred C57BL , Mice, Knockout
20.
Blood ; 124(4): 519-29, 2014 Jul 24.
Article in English | MEDLINE | ID: mdl-24740813

ABSTRACT

We previously showed that immature CD166(+) osteoblasts (OB) promote hematopoietic stem cell (HSC) function. Here, we demonstrate that CD166 is a functional HSC marker that identifies both murine and human long-term repopulating cells. Both murine LSKCD48(-)CD166(+)CD150(+) and LSKCD48(-)CD166(+)CD150(+)CD9(+) cells, as well as human Lin(-)CD34(+)CD38(-)CD49f(+)CD166(+) cells sustained significantly higher levels of chimerism in primary and secondary recipients than CD166(-) cells. CD166(-/-) knockout (KO) LSK cells engrafted poorly in wild-type (WT) recipients and KO bone marrow cells failed to radioprotect lethally irradiated WT recipients. CD166(-/-) hosts supported short-term, but not long-term WT HSC engraftment, confirming that loss of CD166 is detrimental to the competence of the hematopoietic niche. CD166(-/-) mice were significantly more sensitive to hematopoietic stress. Marrow-homed transplanted WT hematopoietic cells lodged closer to the recipient endosteum than CD166(-/-) cells, suggesting that HSC-OB homophilic CD166 interactions are critical for HSC engraftment. STAT3 has 3 binding sites on the CD166 promoter and STAT3 inhibition reduced CD166 expression, suggesting that both CD166 and STAT3 may be functionally coupled and involved in HSC competence. These studies illustrate the significance of CD166 in the identification and engraftment of HSC and in HSC-niche interactions, and suggest that CD166 expression can be modulated to enhance HSC function.


Subject(s)
Activated-Leukocyte Cell Adhesion Molecule/physiology , Biomarkers/metabolism , Bone Marrow Cells/metabolism , Hematopoietic Stem Cells/cytology , Stem Cell Niche/physiology , Animals , Antigens, CD/metabolism , Chromatin Immunoprecipitation , Flow Cytometry , Hematopoietic Stem Cells/physiology , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Receptors, Cell Surface/metabolism , Signaling Lymphocytic Activation Molecule Family Member 1
SELECTION OF CITATIONS
SEARCH DETAIL
...