Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Front Immunol ; 13: 841065, 2022.
Article in English | MEDLINE | ID: mdl-35812447

ABSTRACT

The intestinal immune system and microbiota are emerging as important contributors to the development of metabolic syndrome, but the role of intestinal dendritic cells (DCs) in this context is incompletely understood. BATF3 is a transcription factor essential in the development of mucosal conventional DCs type 1 (cDC1). We show that Batf3-/- mice developed metabolic syndrome and have altered localization of tight junction proteins in intestinal epithelial cells leading to increased intestinal permeability. Treatment with the glycolysis inhibitor 2-deoxy-D-glucose reduced intestinal inflammation and restored barrier function in obese Batf3-/- mice. High-fat diet further enhanced the metabolic phenotype and susceptibility to dextran sulfate sodium colitis in Batf3-/- mice. Antibiotic treatment of Batf3-/- mice prevented metabolic syndrome and impaired intestinal barrier function. Batf3-/- mice have altered IgA-coating of fecal bacteria and displayed microbial dysbiosis marked by decreased obesity protective Akkermansia muciniphila, and Bifidobacterium. Thus, BATF3 protects against metabolic syndrome and preserves intestinal epithelial barrier by maintaining beneficial microbiota.


Subject(s)
Gastrointestinal Microbiome , Metabolic Syndrome , Animals , Gastrointestinal Microbiome/genetics , Homeostasis , Intestines/microbiology , Mice , Mice, Inbred C57BL , Obesity
2.
Mucosal Immunol ; 12(3): 644-655, 2019 05.
Article in English | MEDLINE | ID: mdl-30617301

ABSTRACT

T helper 9 (TH9) cells are important for the development of inflammatory and allergic diseases. The TH9 transcriptional network converges signals from cytokines and antigen presentation but is incompletely understood. Here, we identified TL1A, a member of the TNF superfamily, as a strong inducer of mouse and human TH9 differentiation. Mechanistically, TL1A induced the expression of the transcription factors BATF and BATF3 and facilitated their binding to the Il9 promoter leading to enhanced secretion of IL-9. BATF- and BATF3-deficiencies impaired IL-9 secretion under TH9 and TH9-TL1A-polarizing conditions. In vivo, using a T-cell transfer model, we demonstrated that TL1A promoted IL-9-dependent, TH9 cell-induced intestinal and lung inflammation. Neutralizing IL-9 antibodies attenuated TL1A-driven mucosal inflammation. Batf3-/- TH9-TL1A cells induced reduced inflammation and cytokine expression in vivo compared to WT cells. Our results demonstrate that TL1A promotes TH9 cell differentiation and function and define a role for BATF3 in T-cell-driven mucosal inflammation.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Inflammation/immunology , Inflammatory Bowel Diseases/immunology , Interleukin-9/metabolism , Repressor Proteins/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Animals , Antibodies, Neutralizing/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , Cell Differentiation , Cells, Cultured , Humans , Interleukin-9/genetics , Interleukin-9/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic/genetics , Protein Binding , Repressor Proteins/genetics , Signal Transduction , Tumor Necrosis Factor Ligand Superfamily Member 15/metabolism
3.
Brain Behav Immun ; 67: 163-180, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28860067

ABSTRACT

Osteopontin (OPN), a matricellular immunomodulatory cytokine highly expressed by myelomonocytic cells, is known to regulate immune cell migration, communication, and response to brain injury. Enhanced cerebral recruitment of monocytes achieved through glatiramer acetate (GA) immunization or peripheral blood enrichment with bone marrow (BM)-derived CD115+ monocytes (MoBM) curbs amyloid ß-protein (Aß) neuropathology and preserves cognitive function in murine models of Alzheimer's disease (ADtg mice). To elucidate the beneficial mechanisms of these immunomodulatory approaches in AD, we focused on the potential role of OPN in macrophage-mediated Aß clearance. Here, we found extensive OPN upregulation along with reduction of vascular and parenchymal Aß burden in cortices and hippocampi of GA-immunized ADtg mice. Treatment combining GA with blood-grafted MoBM further increased OPN levels surrounding residual Aß plaques. In brains from AD patients and ADtg mice, OPN was also elevated and predominantly expressed by infiltrating GFP+- or Iba1+-CD45high monocyte-derived macrophages engulfing Aß plaques. Following GA immunization, we detected a significant increase in a subpopulation of inflammatory blood monocytes (CD115+CD11b+Ly6Chigh) expressing OPN, and subsequently, an elevated population of OPN-expressing CD11b+Ly6C+CD45high monocyte/macrophages in the brains of these ADtg mice. Correlogram analyses indicate a strong linear correlation between cerebral OPN levels and macrophage infiltration, as well as a tight inverse relation between OPN and Aß-plaque burden. In vitro studies corroborate in vivo findings by showing that GA directly upregulates OPN expression in BM-derived macrophages (MФBM). Further, OPN promotes a phenotypic shift that is highly phagocytic (increased uptake of Aß fibrils and surface scavenger receptors) and anti-inflammatory (altered cell morphology, reduced iNOS, and elevated IL-10 and Aß-degrading enzyme MMP-9). Inhibition of OPN expression in MФBM, either by siRNA, knockout (KOOPN), or minocycline, impairs uptake of Aß fibrils and hinders GA's neuroprotective effects on macrophage immunological profile. Addition of human recombinant OPN reverses the impaired Aß phagocytosis in KOOPN-MФBM. This study demonstrates that OPN has an essential role in modulating macrophage immunological profile and their ability to resist pathogenic forms of Aß.


Subject(s)
Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Brain/metabolism , Macrophages/immunology , Macrophages/metabolism , Osteopontin/metabolism , Animals , Brain/blood supply , Disease Models, Animal , Encephalitis/metabolism , Female , Male , Mice, Inbred C57BL , Mice, Transgenic , Monocytes/metabolism , Phagocytosis , Up-Regulation
4.
J Leukoc Biol ; 101(3): 727-737, 2017 03.
Article in English | MEDLINE | ID: mdl-27733581

ABSTRACT

TL1A contributes to the pathogenesis of several chronic inflammatory diseases, including those of the bowel by enhancing TH1, TH17, and TH2 responses. TL1A mediates a strong costimulation of these TH subsets, particularly of mucosal CCR9+ T cells. However, the signaling pathways that TL1A induces in different TH subsets are incompletely understood. We investigated the function of TL1A on human TH17 cells. TL1A, together with TGF-ß, IL-6, and IL-23, enhanced the secretion of IL-17 and IFN-γ from human CD4+ memory T cells. TL1A induced expression of the transcription factors BATF and T-bet that correlated with the secretion of IL-17 and IFN-γ. In contrast, TL1A alone induced high levels of IL-22 in memory CD4+ T cells and committed TH17 cells. However, TL1A did not enhance expression of IL-17A in TH17 cells. Expression of the transcription factor aryl hydrocarbon receptor, which regulates the expression of IL-22 was not affected by TL1A. Transcriptome analysis of TH17 cells revealed increased expression of IL-9 in response to TL1A. Blocking IL-9 receptor antibodies abrogated TL1A-induced IL-22 secretion. Furthermore, TL1A increased IL-9 production by peripheral TH17 cells isolated from patients with Crohn's disease. These data suggest that TL1A differentially induces expression of TH17 effector cytokines IL-17, -9, and -22 and provides a potential target for therapeutic intervention in TH17-driven chronic inflammatory diseases.


Subject(s)
Interleukin-9/metabolism , Interleukins/metabolism , Th17 Cells/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 15/metabolism , Cell Separation , Crohn Disease/immunology , Crohn Disease/pathology , Gene Expression Profiling , Humans , Immunologic Memory , Interferon-gamma/metabolism , Leukocyte Common Antigens/metabolism , Interleukin-22
5.
Brain ; 138(Pt 8): 2399-422, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26049087

ABSTRACT

Weekly glatiramer acetate immunization of transgenic mice modelling Alzheimer's disease resulted in retained cognition (Morris water maze test), decreased amyloid-ß plaque burden, and regulation of local inflammation through a mechanism involving enhanced recruitment of monocytes. Ablation of bone marrow-derived myeloid cells exacerbated plaque pathology, whereas weekly administration of glatiramer acetate enhanced cerebral recruitment of innate immune cells, which dampened the pathology. Here, we assessed the therapeutic potential of grafted CD115(+) monocytes, injected once monthly into the peripheral blood of transgenic APPSWE/PS1ΔE9 Alzheimer's disease mouse models, with and without weekly immunization of glatiramer acetate, as compared to glatiramer acetate alone. All immune-modulation treatment groups were compared with age-matched phosphate-buffered saline-injected control transgenic and untreated non-transgenic mouse groups. Two independent cohorts of mice were assessed for behavioural performance (6-8 mice/group); treatments started in 10-month-old symptomatic mice and spanned a total of 2 months. For all three treatments, our data suggest a substantial decrease in cognitive deficit as assessed by the Barnes maze test (P < 0.0001-0.001). Improved cognitive function was associated with synaptic preservation and reduction in cerebral amyloid-ß protein levels and astrogliosis (P < 0.001 and P < 0.0001), with no apparent additive effects for the combined treatment. The peripherally grafted, green fluorescent protein-labelled and endogenous monocytes, homed to cerebral amyloid plaques and directly engulfed amyloid-ß; their recruitment was further enhanced by glatiramer acetate. In glatiramer acetate-immunized mice and, moreover, in the combined treatment group, monocyte recruitment to the brain was coupled with greater elevation of the regulatory cytokine IL10 surrounding amyloid-ß plaques. All treated transgenic mice had increased cerebral levels of MMP9 protein (P < 0.05), an enzyme capable of degrading amyloid-ß, which was highly expressed by the infiltrating monocytes. In vitro studies using primary cultures of bone marrow monocyte-derived macrophages, demonstrated that glatiramer acetate enhanced the ability of macrophages to phagocytose preformed fibrillar amyloid-ß1-42 (P < 0.0001). These glatiramer acetate-treated macrophages exhibited increased expression of the scavenger receptors CD36 and SCARA1 (encoded by MSR1), which can facilitate amyloid-ß phagocytosis, and the amyloid-ß-degrading enzyme MMP9 (P < 0.0001-0.001). Overall, our studies indicate that increased cerebral infiltration of monocytes, either by enrichment of their levels in the circulation or by weekly immunization with glatiramer acetate, resulted in substantial attenuation of disease progression in murine Alzheimer's models by mechanisms that involved enhanced cellular uptake and enzymatic degradation of toxic amyloid-ß as well as regulation of brain inflammation.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Brain/pathology , Monocytes/cytology , Alzheimer Disease/therapy , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Disease Models, Animal , Mice, Transgenic , Microglia/metabolism , Monocytes/metabolism , Plaque, Amyloid/metabolism , Receptor, Macrophage Colony-Stimulating Factor/metabolism
6.
Bangk Med J ; 10: 83-97, 2015 Sep.
Article in English | MEDLINE | ID: mdl-29142857

ABSTRACT

Professor Black and colleagues have been working to improve the quality and sensitivity of imaging in the early detection of conditions from brain tumors to Alzheimer's disease to enhance treatment protocols and patient management. Professor Black et al introduced nanoparticles to improve MRI imaging. These nanoparticles consist of poly (b-L- malic acid (PMLA)) conjugates with monoclonal antibodies ((mAbs)) and Gd-DOTA. These are known as MRI nano-imaging agents (NIA). Most importantly, they can penetrate the endothelial blood-brain barrier (BBB) to reach brain tumors (primary or metastasis). This is effective in cases of brain tumors or breast cancer or other cancers such as lung cancer and gastric cancer having HER2 and/or EGFR positive crossing BBB. By the covalent conjugation of MR contrast (NIA), the MRI virtual biopsy can differentiate brain tumors from infections or other brain pathological conditions. The brain's intrinsic natural fluorescence such as NADH, FAD, lipopigments and porphyrin in the brain tissue can be identified by using time resolved fluorescence spectroscopy (TRFS) which is operated through the use of ultra-short laser. TRFS produces various color bands to differentiate the tumor from normal brain tissue in real time and registers the data on a 3D map. This is significant, as this will provide a greatly improved assessment methodology of tissue type. Consequently, this will potentially result in shorter operation times as well as more satisfactory tumor removal. In the detection of Alzheimer disease, amyloid plaque is deposited in retina tissue (including the RGC, RNFL and inner plexiform layer) which can produce a fluorescence effect by using curcumin as a contrast. This is then shown by human retina amyloid imaging device. Immunotherapies with glatiramer acetate (GA) have been shown to reduce amyloid deposits in brain and retinal AB deposits in mice. The study of advanced imaging technology and techniques including NIA, TRFS and the detection of amyloid plaque in Alzheimer disease are very important approaches to create a new era for diagnostic and therapeutic management of brain tumors and other cancers (HER2 and/or EGFR positive). This pioneering work by Professor Black, and colleagues, gives rise to a new hope for cancer patients for targeted therapy and for immunotherapies in Alzheimer's disease.

7.
J Clin Invest ; 124(3): 1000-12, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24487585

ABSTRACT

Cognitive decline in patients with Alzheimer's disease (AD) is associated with elevated brain levels of amyloid ß protein (Aß), particularly neurotoxic Aß(1-42). Angiotensin-converting enzyme (ACE) can degrade Aß(1-42), and ACE overexpression in myelomonocytic cells enhances their immune function. To examine the effect of targeted ACE overexpression on AD, we crossed ACE(10/10) mice, which overexpress ACE in myelomonocytes using the c-fms promoter, with the transgenic APP(SWE)/PS1(ΔE9) mouse model of AD (AD⁺). Evaluation of brain tissue from these AD⁺ACE(10/10) mice at 7 and 13 months revealed that levels of both soluble and insoluble brain Aß(1-42) were reduced compared with those in AD⁺ mice. Furthermore, both plaque burden and astrogliosis were drastically reduced. Administration of the ACE inhibitor ramipril increased Aß levels in AD⁺ACE(10/10) mice compared with the levels induced by the ACE-independent vasodilator hydralazine. Overall, AD⁺ACE(10/10) mice had less brain-infiltrating cells, consistent with reduced AD-associated pathology, though ACE-overexpressing macrophages were abundant around and engulfing Aß plaques. At 11 and 12 months of age, the AD⁺ACE(10/WT) and AD⁺ACE(10/10) mice were virtually equivalent to non-AD mice in cognitive ability, as assessed by maze-based behavioral tests. Our data demonstrate that an enhanced immune response, coupled with increased myelomonocytic expression of catalytically active ACE, prevents cognitive decline in a murine model of AD.


Subject(s)
Alzheimer Disease/enzymology , Amyloid beta-Peptides/metabolism , Myeloid Cells/enzymology , Peptidyl-Dipeptidase A/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Astrocytes/physiology , Cell Movement , Cerebral Cortex/blood supply , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cognition , Female , Humans , Macrophages/physiology , Male , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Transgenic , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Ramipril/pharmacology , Solubility
8.
Cancer Metab ; 2: 21, 2014.
Article in English | MEDLINE | ID: mdl-25671108

ABSTRACT

BACKGROUND: Succinate dehydrogenase (SDH) is a mitochondrial metabolic enzyme complex involved in both the electron transport chain and the citric acid cycle. SDH mutations resulting in enzymatic dysfunction have been found to be a predisposing factor in various hereditary cancers. Therefore, SDH has been implicated as a tumor suppressor. RESULTS: We identified that dysregulation of SDH components also occurs in serous ovarian cancer, particularly the SDH subunit SDHB. Targeted knockdown of Sdhb in mouse ovarian cancer cells resulted in enhanced proliferation and an epithelial-to-mesenchymal transition (EMT). Bioinformatics analysis revealed that decreased SDHB expression leads to a transcriptional upregulation of genes involved in metabolic networks affecting histone methylation. We confirmed that Sdhb knockdown leads to a hypermethylated epigenome that is sufficient to promote EMT. Metabolically, the loss of Sdhb resulted in reprogrammed carbon source utilization and mitochondrial dysfunction. This altered metabolic state of Sdhb knockdown cells rendered them hypersensitive to energy stress. CONCLUSIONS: These data illustrate how SDH dysfunction alters the epigenetic and metabolic landscape in ovarian cancer. By analyzing the involvement of this enzyme in transcriptional and metabolic networks, we find a metabolic Achilles' heel that can be exploited therapeutically. Analyses of this type provide an understanding how specific perturbations in cancer metabolism may lead to novel anticancer strategies.

9.
Neurodegener Dis ; 10(1-4): 285-93, 2012.
Article in English | MEDLINE | ID: mdl-22343730

ABSTRACT

BACKGROUND: Definite Alzheimer's disease (AD) diagnosis at early stages is vital for targeting intervention, yet currently unavailable. Noninvasive detection of the pathological hallmark, amyloid-ß protein (Aß) plaques, is limited in the brain. However, the existence of Aß plaques in the retina, possibly at presymptomatic stages, may improve early detection of AD. OBJECTIVE: To summarize clinical and preclinical evidence showing that the retina, an accessible part of the central nervous system, displays abnormalities in AD, especially Aß plaque pathology. The ability to monitor in vivo retinal plaque dynamics in response to immunotherapy is also assessed. METHODS: Literature analysis of retinal AD pathology and imaging is provided. In our studies, systemic curcumin is administered to enable monitoring of retinal Aß plaques in live APP(SWE)/PS1(Δ)(E9) transgenic mice by optical imaging. RESULTS: Visual and retinal abnormalities, including early manifestation of retinal Aß plaque pathology, have been documented in AD patients and animal models. In mouse models, retinal Aß plaques accumulate with age and decrease in response to immunotherapy, consistent with brain pathology. Here, we demonstrate that retinal plaques can be individually monitored in real time following glatiramer acetate immunization. CONCLUSION: Translation of noninvasive retinal-plaque imaging to humans could eventually facilitate early and accurate AD diagnosis and therapy assessment.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Retina/drug effects , Retina/metabolism , Alzheimer Disease/complications , Alzheimer Disease/drug therapy , Amyloid beta-Protein Precursor/genetics , Animals , Curcumin/therapeutic use , Disease Models, Animal , Early Diagnosis , Humans , Mice , Mice, Transgenic , Neuroimaging/methods , Presenilin-1/genetics , Vision Disorders/etiology , Vision Disorders/genetics , Vision Disorders/pathology
10.
Invest Ophthalmol Vis Sci ; 52(12): 9033-46, 2011 Nov 21.
Article in English | MEDLINE | ID: mdl-21969301

ABSTRACT

PURPOSE: Immunization with glatiramer acetate (GA) alleviates the neuropathology associated with glaucoma and Alzheimer's disease (AD) in rodent models. This research was undertaken to screen for molecular factors underlying GA-induced neuroprotective mechanisms in these models of chronic neurodegeneration. METHODS: Gene expression profiles were analyzed in GA-immunized versus nonimmunized elevated-intraocular pressure (IOP) rat models of glaucoma by using whole genome cDNA microarrays and were further validated by quantitative real-time PCR analysis. A gene, prominently upregulated by GA in elevated IOP retina, was further studied in APP(SWE)/PS1(ΔE9)-transgenic (AD-Tg) mice after GA immunization. RESULTS: Seven days after treatment with GA, numerous genes were regulated in the retinas of rats with elevated IOP. Comprehensive functional classification and DAVID/KEGG enrichment analysis of GA-induced differentially expressed genes revealed annotation terms and pathways involved in neuroprotection, immune responses, cell communication, and regeneration. Specifically, increased mRNA levels of an early growth response (Egr) 1 gene were evident in GA-immunized retinas with elevated IOP. In AD-Tg mice, a significant increase in hippocampal EGR1 protein levels was also found in response to GA immunization. Nuclear EGR1 in the dentate gyrus colocalized more frequently with doublecortin-positive and Ki67 proliferating neural progenitors in GA-immunized as compared to nonimmunized AD-Tg mice. Further, EGR1 levels were negatively correlated with hippocampal amyloid-ß plaque burden. CONCLUSIONS: This study presents global gene expression profiles associated with GA immunization in a glaucoma rat model. Moreover, it identifies EGR1 transcription factor as a potential mediator for GA-induced neuroprotection in both glaucoma and AD.


Subject(s)
Alzheimer Disease/drug therapy , Early Growth Response Protein 1/genetics , Glaucoma/drug therapy , Immunosuppressive Agents/pharmacology , Peptides/pharmacology , Alzheimer Disease/genetics , Alzheimer Disease/immunology , Animals , Chronic Disease , Disease Models, Animal , Doublecortin Protein , Female , Gene Expression/drug effects , Gene Expression/immunology , Glatiramer Acetate , Glaucoma/genetics , Glaucoma/immunology , Hippocampus/physiology , Male , Mice , Mice, Transgenic , Nerve Degeneration/drug therapy , Nerve Degeneration/genetics , Nerve Degeneration/immunology , Neuroprotective Agents/pharmacology , Oligonucleotide Array Sequence Analysis , Rats , Rats, Inbred Lew , Real-Time Polymerase Chain Reaction
11.
Mol Cancer Ther ; 9(6): 1525-35, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20501804

ABSTRACT

Tyrosine kinase inhibitors (TKI) targeting angiogenesis via inhibition of the vascular endothelial growth factor pathway have changed the medical management of metastatic renal cell carcinoma. Although treatment with TKIs has shown clinical benefit, these drugs will eventually fail patients. The potential mechanisms of resistance to TKIs are poorly understood. To address this question, we obtained an excisional biopsy of a skin metastasis from a patient with clear cell renal carcinoma who initially had a response to sunitinib and eventually progressed with therapy. Tumor pieces were grafted s.c. in athymic nude mice. Established xenografts were treated with sunitinib. Tumor size, microvascular density, and pericyte coverage were determined. Plasma as well as tissue levels for sunitinib were assessed. A tumor-derived cell line was established and assessed in vitro for potential direct antitumor effects of sunitinib. To our surprise, xenografts from the patient who progressed on sunitinib regained sensitivity to the drug. At a dose of 40 mg/kg, sunitinib caused regression of the subcutaneous tumors. Histology showed a marked reduction in microvascular density and pericyte dysfunction. More interestingly, histologic examination of the original skin metastasis revealed evidence of epithelial to mesenchymal transition, whereas the xenografts showed reversion to the clear cell phenotype. In vitro studies showed no inhibitory effect on tumor cell growth at pharmacologically relevant concentrations. In conclusion, the histologic examination in this xenograft study suggests that reversible epithelial to mesenchymal transition may be associated with acquired tumor resistance to TKIs in patients with clear cell renal carcinoma.


Subject(s)
Carcinoma, Renal Cell/drug therapy , Drug Resistance, Neoplasm , Epithelium/pathology , Indoles/therapeutic use , Kidney Neoplasms/drug therapy , Mesoderm/pathology , Pyrroles/therapeutic use , Xenograft Model Antitumor Assays , Angiogenesis Inhibitors/blood , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Disease Progression , Drug Resistance, Neoplasm/drug effects , Epithelium/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Indoles/blood , Indoles/pharmacology , Kidney Neoplasms/blood supply , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Mesoderm/drug effects , Mice , Mice, Nude , Microvessels/drug effects , Pericytes/drug effects , Pericytes/pathology , Phenotype , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyrroles/blood , Pyrroles/pharmacology , Skin Neoplasms/secondary , Sunitinib , Von Hippel-Lindau Tumor Suppressor Protein/genetics
12.
Curr Protoc Pharmacol ; Chapter 14: Unit 14.13, 2009 Dec.
Article in English | MEDLINE | ID: mdl-22294393

ABSTRACT

Significant progress has been made in the treatment of patients with advanced renal cancer. In addition to immunotherapy, there are several potentially distinct therapeutic approaches for targeting molecular pathways. The murine models detailed in this unit are useful for testing rational combination strategies. Moreover, animal models contribute immensely to the understanding of the genetic, epigenetic, and biological aspects of human disease. Compared to humans, rodent models are relatively short-lived and allow for the facile study of clinically relevant pathologies. Animal models for the study of renal cell carcinoma (RCC) are particularly useful for the development of new drugs for kidney cancer. Included in this unit are several in vivo models that are currently used to evaluate therapeutic approaches to renal cancer therapy and to investigate the pathophysiology of this condition. Included are both murine (RENCA) and renal cell carcinomas in subcutaneous and orthotopic models using tumor cell lines and human tumor tissue.


Subject(s)
Adenocarcinoma/drug therapy , Carcinoma, Renal Cell/drug therapy , Disease Models, Animal , Kidney Neoplasms/drug therapy , Animals , Cell Line, Tumor , Drug Discovery/methods , Drug Evaluation, Preclinical , Drug Screening Assays, Antitumor/methods , Humans , Indicators and Reagents , Luminescent Measurements , Mice , Mice, Inbred BALB C , Neoplasm Transplantation/methods , Solutions
13.
Clin Cancer Res ; 14(11): 3589-97, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18519793

ABSTRACT

PURPOSE: The hypoxia-inducible factor-1 alpha (HIF-alpha) is a key regulator of tumor angiogenesis. Mammalian target of rapamycin (mTOR) and histone deacetylase (HDAC) inhibitors suppress tumor-induced angiogenesis by reducing tumor HIF-1 alpha protein expression. Thus, we hypothesized that combination treatment of rapamycin and the HDAC inhibitor LBH589 has greater antiangiogenic and antitumor activity compared with single agents. EXPERIMENTAL DESIGN: To evaluate the effect of LBH589 and rapamycin on HIF-1 alpha in human prostate PC3, renal C2 carcinoma cell lines, and endothelial cells (human umbilical vein endothelial cells), we did Western blot analysis. To determine the antitumor activity of LBH589 and rapamycin, cell proliferation assays and xenograft experiments were conducted. RESULTS: Western blotting showed that combination treatment of human umbilical vein endothelial cells, C2 and PC3, significantly reduced HIF-1 alpha protein expression compared with single agents. Treatment with rapamycin resulted in inhibition of the downstream signals of the mTOR pathway and increased phosphorylation of Akt in C2 cells, whereas the constitutively activated Akt in PC3 cells was not modulated. LBH589 decreased both constitutively expressed and rapamycin-induced phosphorylated Akt levels in PC3 and C2 cell lines. In clonogenic assays, the combination treatment had a greater inhibitory effect in PC3 cells (93 +/- 1.4%) compared with single agents (66 +/- 9% rapamycin and 43 +/- 4% LBH589). Combination of rapamycin and LBH589 significantly inhibited PC3 and C2 in vivo tumor growth and angiogenesis as measured by tumor weight and microvessel density. CONCLUSIONS: Combination treatment of mTOR and HDAC inhibitors represents a rational therapeutic strategy targeting HIF-1 alpha that warrants clinical testing.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Hydroxamic Acids/administration & dosage , Neoplasms, Experimental/drug therapy , Sirolimus/administration & dosage , Blotting, Western , Cell Line, Tumor , Endothelial Cells/drug effects , Enzyme Inhibitors/administration & dosage , Histone Deacetylases/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/drug effects , Immunohistochemistry , Indoles , Neovascularization, Pathologic/drug therapy , Oncogene Protein v-akt/drug effects , Oncogene Protein v-akt/metabolism , Panobinostat , Phosphorylation/drug effects , Protein Kinases/drug effects , Signal Transduction/drug effects , TOR Serine-Threonine Kinases , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/drug effects , Xenograft Model Antitumor Assays
14.
Clin Cancer Res ; 13(15 Pt 1): 4538-46, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17671140

ABSTRACT

PURPOSE: High-dose interleukin 2 (IL-2) is a Food and Drug Administration-approved regimen for patients with metastatic renal cell carcinoma. However, the toxicity and limited clinical benefit associated with IL-2 has hampered its use. Histone deacetylase (HDAC) inhibitors have been shown to have antitumor activity in different tumor models including renal cell carcinoma, and to have immunomodulatory properties. In our study, we tested the effectiveness of combination therapy of IL-2 with the HDAC inhibitor MS-275 in a murine renal cell carcinoma (RENCA) model. EXPERIMENTAL DESIGN: RENCA luciferase-expressing cells were implanted in the left kidney of BALB/C mice. Animals were randomly divided into four groups and treated with either vehicle, 150,000 IU of IL-2 twice daily by i.p. injections (twice weekly), 5 mg/kg of MS-275 daily by oral gavage (5 d/wk), or its combination. Treatment was started either 3 or 9 days following tumor cell injection. RESULTS: Weekly luciferase images and tumor weight after 2 weeks of treatment showed significant tumor inhibition (>80%) in the combination treatment as compared with the IL-2 (no significant inhibition) or MS-275 (approximately 40% inhibition) treatment groups. Spontaneous lung metastases were also inhibited in the combination treatment (>90% inhibition) as compared with the single treatment group. Kaplan-Meier analyses showed statistically significant increased survival in the combination group as compared with controls and single agents. Splenocytes from mice treated with combination treatment showed greater lysis of RENCA cells than splenocytes from mice treated with single agents. The percentage of CD4(+)CD25(+) T cells and Foxp3(+) T cells (T regulatory cells) was increased or reduced, respectively, in lymph nodes from tumor-bearing animals treated with the combination of MS-275 and IL-2 as compared with control and single agents. Depletion of CD8(+) T cells abrogated the survival benefit from MS-275 + IL-2 combination. CONCLUSIONS: These results show that the combination of IL-2 and MS-275 has a synergistic antitumor effect in vivo in an immunocompetent murine model of renal cell carcinoma. The antitumor effect was associated with the decreased number of T regulatory cells and the increased antitumor cytotoxicity by splenocytes. In conclusion, these preclinical data provide the rationale for clinical testing of the combination of IL-2 and HDAC inhibitors in the treatment of patients with renal cell carcinoma.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzamides/therapeutic use , Carcinoma, Renal Cell/drug therapy , Histone Deacetylase Inhibitors , Interleukin-2/therapeutic use , Kidney Neoplasms/drug therapy , Pyridines/therapeutic use , Animals , Cell Proliferation/drug effects , Drug Synergism , Drug Therapy, Combination , Female , Flow Cytometry , Mice , Mice, Inbred BALB C , Mice, Nude , Survival Rate
15.
Clin Cancer Res ; 13(14): 4201-8, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17634549

ABSTRACT

PURPOSE: Angiogenesis inhibitors have shown clinical benefit in patients with advanced renal cell cancer, but further therapeutic improvement is needed. Vascular endothelial growth factor (VEGF) Trap is a newly developed VEGF-blocking agent with stronger affinity and broader activity than the anti-VEGF antibody bevacizumab. In this study, we tested the activity of VEGF Trap in an orthotopic murine model of renal cancer with spontaneous lung metastases. EXPERIMENTAL DESIGN: Murine syngeneic renal cell carcinoma cells (RENCA) transfected with a luciferase-expressing vector were injected into the renal capsule of BALB/c mice. I.p. treatment with VEGF Trap or control protein (10 or 25 mg/kg twice weekly) was started shortly after tumor injection to prevent tumor development (prevention model) or after established tumors were formed to inhibit tumor growth and metastasis formation (intervention model). RESULTS: In the prevention model, VEGF Trap inhibited tumor growth by 87 +/- 14% compared with control (P=0.007) and significantly prolonged survival. In the intervention model, VEGF Trap inhibited tumor growth by 74 +/- 9% (P<0.001) and the formation of lung metastases was inhibited by 98% (P<0.004). Microvascular density was reduced by 66% due to VEGF Trap treatment (P<0.001). In addition, VEGF Trap prevented fibrinogen leakage into the tumor microenvironment representative for reduced vascular leaking as shown by immunohistochemical staining. CONCLUSIONS: VEGF Trap is a potent inhibitor of RENCA tumor growth and metastasis formation and blocks the biological function of VEGF in vivo. These results support further clinical development of VEGF Trap for renal cell cancer and other cancer types.


Subject(s)
Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , Neovascularization, Pathologic/prevention & control , Recombinant Fusion Proteins/therapeutic use , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Carcinoma, Renal Cell/blood supply , Cell Line, Tumor , Disease Models, Animal , Kidney Neoplasms/blood supply , Mice , Microcirculation/drug effects , Microcirculation/pathology , Pericytes/drug effects , Pericytes/pathology
16.
Mol Cancer Ther ; 6(1): 70-81, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17237267

ABSTRACT

Resistance to chemotherapy is a major hurdle in the treatment of malignant melanoma. Histone deacetylase (HDAC) inhibitors have been shown to have antitumor activity in different tumor types, including melanoma, and to reverse epigenetic repression of tumor suppressor genes, such as retinoic acid receptor beta (RARbeta). In this study, we tested the antitumor effect of the HDAC inhibitor LAQ824 in combination with 13-cis-retinoic acid (CRA) on two human melanoma cell lines both in vitro and in vivo. Treatment of LAQ824 showed a dose-dependent inhibitory effect on A2058 and HMV-I cell lines in a clonogenic assay. These cell lines were relatively resistance to CRA. On treatment with combination of LAQ824 and CRA, a greater inhibitory effect (up to 98%) was achieved compared with single agents. Lack of RARbeta2 gene expression was associated with histone acetylation and gene methylation at the promoter level. Treatment with LAQ824 restored retinoid sensitivity by reverting RARbeta2 epigenetic silencing. The biological effect of LAQ824 was associated with p21 induction in both cell lines but G(2) cell cycle arrest in A2058 and apoptosis in HMV-I cell line. The induction of apoptosis by LAQ824 was associated with increased reactive oxygen species and induction of SM22 gene expression in HMV-I but not in A2058 cell line. Administration of the free radical scavenger l-N-acetylcysteine blocked LAQ824 + CRA-mediated apoptosis in HMV-I cells, suggesting a primary role for reactive oxygen species generation in LAQ824 + CRA-associated lethality. Combination treatment showed 61% and 82% growth inhibition in A2058 and HMV-I tumors, respectively. Greater induction of in vivo apoptosis was observed in the HMV-I but not in the A2058 tumors treated with combination therapy compared with single agents. These results suggest that the HDAC inhibitor LAQ824 has a greater antitumor activity in combination with CRA in melanoma tumors but the degree of induced apoptosis may vary. Combination of HDAC inhibitors and retinoids represents a novel therapeutic approach for malignant melanoma that warrants clinical testing.


Subject(s)
Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Isotretinoin/pharmacology , Melanoma/drug therapy , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , G2 Phase/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hydroxamic Acids/administration & dosage , Hydroxamic Acids/therapeutic use , Isotretinoin/administration & dosage , Isotretinoin/therapeutic use , Male , Melanoma/genetics , Melanoma/pathology , Mice , Mice, Nude , Microfilament Proteins/genetics , Muscle Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Receptors, Retinoic Acid/genetics
17.
Clin Cancer Res ; 12(2): 634-42, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16428510

ABSTRACT

PURPOSE: Angiogenesis is required for tumor progression and represents a rational target for therapeutic intervention. Histone deacetylase (HDAC) inhibitors have been shown to have activity against various tumor cell types by inhibiting proliferation and inducing apoptosis both in vitro and in vivo. HDAC inhibitors have also been reported to inhibit angiogenesis. The goal of this study was to characterize the antiangiogenic and antitumor activity of a recently developed HDAC inhibitor, the hydroxamic derivative LBH589. MATERIALS AND METHODS: To evaluate the antiangiogenesis activity of LBH589, we did cell cycle analysis, cell proliferation, tube formation, invasion assays in vitro, and Matrigel plug assay in vivo. To determine the antitumor activity of LBH589, we established human prostate carcinoma cell PC-3 xenografts in vivo. To evaluate the effect of LBH589 on endothelial signaling pathways, gene expression, and protein acetylation, we did Western blots and reverse transcription-PCR in human umbilical vein endothelial cells (HUVEC). Immunohistochemical analysis was done to evaluate new blood vessel formation in vivo. RESULTS: LBH589 induced acetylation of histone H3 and alpha-tubulin protein in HUVECs. Histone and nonhistone protein acetylation correlated with induction of G(2)-M cell cycle arrest, inhibition of HUVEC proliferation, and viability. Noncytotoxic concentrations of LBH589 inhibited endothelial tube formation, Matrigel invasion, AKT, extracellular signal-regulated kinase 1/2 phosphorylation, and chemokine receptor CXCR4 expression. In vivo dosing of mice with LBH589 (10 mg/kg/d) reduced angiogenesis and PC-3 tumor growth. CONCLUSION: This study provides evidence that LBH589 induces a wide range of effects on endothelial cells that lead to inhibition of tumor angiogenesis. These results support the role of HDAC inhibitors as a therapeutic strategy to target both the tumor and endothelial compartment and warrant the clinical development of these agents in combination with angiogenesis inhibitors.


Subject(s)
Cell Division/drug effects , Cell Proliferation/drug effects , G2 Phase/drug effects , Neovascularization, Pathologic/drug therapy , Prostatic Neoplasms/drug therapy , Acetylation/drug effects , Angiogenesis Inhibitors/pharmacology , Animals , Blotting, Western , Collagen/metabolism , Drug Combinations , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Histones/metabolism , Humans , Hydroxamic Acids/therapeutic use , Indoles , Laminin/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasm Invasiveness , Panobinostat , Phosphorylation/drug effects , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proteoglycans/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, CXCR4/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Transplantation, Heterologous , Tubulin/metabolism , Umbilical Veins/cytology , Umbilical Veins/drug effects , Umbilical Veins/metabolism , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...