Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Nat Cancer ; 5(1): 85-99, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37814010

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) cells use glutamine (Gln) to support proliferation and redox balance. Early attempts to inhibit Gln metabolism using glutaminase inhibitors resulted in rapid metabolic reprogramming and therapeutic resistance. Here, we demonstrated that treating PDAC cells with a Gln antagonist, 6-diazo-5-oxo-L-norleucine (DON), led to a metabolic crisis in vitro. In addition, we observed a profound decrease in tumor growth in several in vivo models using sirpiglenastat (DRP-104), a pro-drug version of DON that was designed to circumvent DON-associated toxicity. We found that extracellular signal-regulated kinase (ERK) signaling is increased as a compensatory mechanism. Combinatorial treatment with DRP-104 and trametinib led to a significant increase in survival in a syngeneic model of PDAC. These proof-of-concept studies suggested that broadly targeting Gln metabolism could provide a therapeutic avenue for PDAC. The combination with an ERK signaling pathway inhibitor could further improve the therapeutic outcome.


Subject(s)
Antineoplastic Agents , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Glutamine/metabolism , Cell Line, Tumor , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Antineoplastic Agents/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/metabolism , Enzyme Inhibitors/pharmacology
2.
Sci Adv ; 9(16): eadf9284, 2023 04 21.
Article in English | MEDLINE | ID: mdl-37075122

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) cells maintain a high level of autophagy, allowing them to thrive in an austere microenvironment. However, the processes through which autophagy promotes PDAC growth and survival are still not fully understood. Here, we show that autophagy inhibition in PDAC alters mitochondrial function by losing succinate dehydrogenase complex iron sulfur subunit B expression by limiting the availability of the labile iron pool. PDAC uses autophagy to maintain iron homeostasis, while other tumor types assessed require macropinocytosis, with autophagy being dispensable. We observed that cancer-associated fibroblasts can provide bioavailable iron to PDAC cells, promoting resistance to autophagy ablation. To overcome this cross-talk, we used a low-iron diet and demonstrated that this augmented the response to autophagy inhibition therapy in PDAC-bearing mice. Our work highlights a critical link between autophagy, iron metabolism, and mitochondrial function that may have implications for PDAC progression.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Animals , Mice , Cell Line, Tumor , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/metabolism , Autophagy , Homeostasis , Mitochondria/metabolism , Tumor Microenvironment , Pancreatic Neoplasms
3.
Nat Commun ; 12(1): 4905, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34385458

ABSTRACT

α-ketoglutarate (KG), also referred to as 2-oxoglutarate, is a key intermediate of cellular metabolism with pleiotropic functions. Cell-permeable esterified analogs are widely used to study how KG fuels bioenergetic and amino acid metabolism and DNA, RNA, and protein hydroxylation reactions, as cellular membranes are thought to be impermeable to KG. Here we show that esterified KG analogs rapidly hydrolyze in aqueous media, yielding KG that, in contrast to prevailing assumptions, imports into many cell lines. Esterified KG analogs exhibit spurious KG-independent effects on cellular metabolism, including extracellular acidification, arising from rapid hydrolysis and de-protonation of α-ketoesters, and significant analog-specific inhibitory effects on glycolysis or mitochondrial respiration. We observe that imported KG decarboxylates to succinate in the cytosol and contributes minimally to mitochondrial metabolism in many cell lines cultured in normal conditions. These findings demonstrate that nuclear and cytosolic KG-dependent reactions may derive KG from functionally distinct subcellular pools and sources.


Subject(s)
Amino Acids/metabolism , Energy Metabolism , Esters/metabolism , Ketoglutaric Acids/metabolism , Mitochondria/metabolism , Succinic Acid/metabolism , Animals , Cell Line, Tumor , Cytosol/metabolism , Esters/chemistry , Glycolysis , HEK293 Cells , Humans , Hydrolysis , Hydrophobic and Hydrophilic Interactions , Ketoglutaric Acids/chemistry , Mice , Oxygen Consumption , RAW 264.7 Cells
4.
Nature ; 581(7806): 100-105, 2020 05.
Article in English | MEDLINE | ID: mdl-32376951

ABSTRACT

Immune evasion is a major obstacle for cancer treatment. Common mechanisms of evasion include impaired antigen presentation caused by mutations or loss of heterozygosity of the major histocompatibility complex class I (MHC-I), which has been implicated in resistance to immune checkpoint blockade (ICB) therapy1-3. However, in pancreatic ductal adenocarcinoma (PDAC), which is resistant to most therapies including ICB4, mutations that cause loss of MHC-I are rarely found5 despite the frequent downregulation of MHC-I expression6-8. Here we show that, in PDAC, MHC-I molecules are selectively targeted for lysosomal degradation by an autophagy-dependent mechanism that involves the autophagy cargo receptor NBR1. PDAC cells display reduced expression of MHC-I at the cell surface and instead demonstrate predominant localization within autophagosomes and lysosomes. Notably, inhibition of autophagy restores surface levels of MHC-I and leads to improved antigen presentation, enhanced anti-tumour T cell responses and reduced tumour growth in syngeneic host mice. Accordingly, the anti-tumour effects of autophagy inhibition are reversed by depleting CD8+ T cells or reducing surface expression of MHC-I. Inhibition of autophagy, either genetically or pharmacologically with chloroquine, synergizes with dual ICB therapy (anti-PD1 and anti-CTLA4 antibodies), and leads to an enhanced anti-tumour immune response. Our findings demonstrate a role for enhanced autophagy or lysosome function in immune evasion by selective targeting of MHC-I molecules for degradation, and provide a rationale for the combination of autophagy inhibition and dual ICB therapy as a therapeutic strategy against PDAC.


Subject(s)
Adenocarcinoma/immunology , Autophagy/immunology , Carcinoma, Pancreatic Ductal/immunology , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Pancreatic Neoplasms/immunology , Tumor Escape/immunology , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Antigen Presentation/drug effects , Antigen Presentation/immunology , Autophagy/drug effects , Autophagy/genetics , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/immunology , Cell Line, Tumor , Chloroquine/pharmacology , Female , Histocompatibility Antigens Class I/genetics , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Male , Mice , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Tumor Escape/drug effects
5.
Cancer Cell ; 35(4): 588-602.e10, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30930117

ABSTRACT

The roles of tumor-associated macrophages (TAMs) and circulating monocytes in human cancer are poorly understood. Here, we show that monocyte subpopulation distribution and transcriptomes are significantly altered by the presence of endometrial and breast cancer. Furthermore, TAMs from endometrial and breast cancers are transcriptionally distinct from monocytes and their respective tissue-resident macrophages. We identified a breast TAM signature that is highly enriched in aggressive breast cancer subtypes and associated with shorter disease-specific survival. We also identified an auto-regulatory loop between TAMs and cancer cells driven by tumor necrosis factor alpha involving SIGLEC1 and CCL8, which is self-reinforcing through the production of CSF1. Together these data provide direct evidence that monocyte and macrophage transcriptional landscapes are perturbed by cancer, reflecting patient outcomes.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Cellular Reprogramming , Macrophages/metabolism , Monocytes/metabolism , Paracrine Communication , Transcription, Genetic , Antineoplastic Agents/pharmacology , Biomarkers, Tumor/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Chemokine CCL8/genetics , Chemokine CCL8/metabolism , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Induced Pluripotent Stem Cells/metabolism , Macrophage Colony-Stimulating Factor/genetics , Macrophages/pathology , Molecular Targeted Therapy , Monocytes/pathology , Sialic Acid Binding Ig-like Lectin 1/genetics , Sialic Acid Binding Ig-like Lectin 1/metabolism , Signal Transduction , THP-1 Cells , Tumor Microenvironment
6.
Cancer Discov ; 8(3): 276-287, 2018 03.
Article in English | MEDLINE | ID: mdl-29317452

ABSTRACT

Autophagy has been shown to be elevated in pancreatic ductal adenocarcinoma (PDAC), and its role in promoting established tumor growth has made it a promising therapeutic target. However, due to limitations of prior mouse models as well as the lack of potent and selective autophagy inhibitors, the ability to fully assess the mechanistic basis of how autophagy supports pancreatic cancer has been limited. To test the feasibility of treating PDAC using autophagy inhibition and further our understanding of the mechanisms of protumor effects of autophagy, we developed a mouse model that allowed the acute and reversible inhibition of autophagy. We observed that autophagy inhibition causes significant tumor regression in an autochthonous mouse model of PDAC. A detailed analysis of these effects indicated that the tumor regression was likely multifactorial, involving both tumor cell-intrinsic and host effects. Thus, our study supports that autophagy inhibition in PDAC may have future utility in the treatment of pancreatic cancer and illustrates the importance of assessing complex biological processes in relevant autochthonous models.Significance: This work demonstrates that autophagy is critical pancreatic tumor maintenance through tumor cell-intrinsic and -extrinsic mechanisms. These results have direct clinical relevance to ongoing clinical trials as well as drug-development initiatives. Cancer Discov; 8(3); 276-87. ©2018 AACR.See related commentary by Noguera-Ortega and Amaravadi, p. 266This article is highlighted in the In This Issue feature, p. 253.


Subject(s)
Autophagy/physiology , Carcinoma, Pancreatic Ductal/pathology , Neoplasms, Experimental/genetics , Pancreatic Neoplasms/pathology , Animals , Autophagy-Related Proteins/genetics , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Cysteine Endopeptidases/genetics , Doxycycline/pharmacology , Mice, Transgenic , Neoplasms, Experimental/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics
7.
Congenit Heart Dis ; 10(5): 387-95, 2015.
Article in English | MEDLINE | ID: mdl-25358483

ABSTRACT

BACKGROUND: Adults with congenital heart disease (CHD) face increased risk for morbidity and mortality with age, but few prognostic models exist. OBJECTIVE: This study aims to assess whether the Heart Failure Survival Score (HFSS), which risk stratifies patients for heart transplantation, predicts outcomes in adults with moderate or complex CHD. METHODS: This was a multicenter, retrospective study which identified 441 patients with moderate or complex CHD between 2005 and 2013, of whom 169 had all the HFSS parameters required to calculate the risk score. Because all study patients were deemed low risk by the HFSS, the score was dichotomized at the median (10.4). Outcomes included death, transplant or ventricular assist device (VAD), arrhythmia requiring treatment, nonelective cardiovascular (CV) hospitalizations, and the composite. Associations of mean HFSS and HFSS <10.4 with each outcome were assessed. RESULTS: The cohort had mean ± standard deviation age of 33.6 ± 12.6 years, peak VO2 21.8 ± 7.5 mL/kg/min, HFSS of 10.45 ± 0.88, and median years follow-up of 2.7 (1.1, 5.2). There were five deaths (2.8%), no transplants or VADs, 25 arrhythmias (14.8%), 22 CV hospitalizations (13%), and 39 composites (23.1%). Lower mean HFSS was observed for patients who died (9.6 ± 0.83 vs. 10.5 ± 0.87, P = .02), arrhythmia requiring treatment (10.0 ± 0.70 vs. 10.5 ± 0.89, P = .005), CV hospitalizations (9.9 ± 0.73 vs. 10.5 ± 0.88, P = .002), and the composite (10.0 ± 0.70 vs. 10.6 ± 0.89, P < .001). The positive and negative predictive values of HFSS <10.4 for the composite were 34% and 88% respectively, with sensitivity and specificity 74% and 56%. CONCLUSIONS: Although a low HFSS was significantly associated with outcomes, it did not adequately risk stratify adults with CHD, whose heterogeneous pathophysiology differs from that of the acquired heart failure population. Further studies are warranted to provide a more accurate prognosis.


Subject(s)
Decision Support Techniques , Heart Defects, Congenital/complications , Heart Failure/etiology , Adult , Age Factors , Boston , Female , Heart Defects, Congenital/diagnosis , Heart Defects, Congenital/mortality , Heart Defects, Congenital/surgery , Heart Failure/diagnosis , Heart Failure/mortality , Heart Failure/surgery , Heart Transplantation , Hospitalization , Humans , Male , Middle Aged , New York , Predictive Value of Tests , Prognosis , Retrospective Studies , Risk Assessment , Risk Factors , Time Factors , Young Adult
8.
Am J Pathol ; 184(12): 3359-75, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25451155

ABSTRACT

Progression to an angiogenic state is a critical event in tumor development, yet few patient characteristics have been identified that can be mechanistically linked to this transition. Antiphospholipid autoantibodies (aPLs) are prevalent in many human cancers and can elicit proangiogenic expression in several cell types, but their role in tumor biology is unknown. Herein, we observed that the elevation of circulating aPLs among breast cancer patients is specifically associated with invasive-stage tumors. By using multiple in vivo models of breast cancer, we demonstrated that aPL-positive IgG from patients with autoimmune disease rapidly accelerates tumor angiogenesis and consequent tumor progression, particularly in slow-growing avascular tumors. The action of aPLs was local to the tumor site and elicited leukocytic infiltration and tumor invasion. Tumor cells treated with aPL-positive IgG expressed multiple proangiogenic genes, including vascular endothelial growth factor, tissue factor (TF), and colony-stimulating factor 1. Knockdown and neutralization studies demonstrated that the effects of aPLs on tumor angiogenesis and growth were dependent on tumor cell-derived TF. Tumor-derived TF was essential for the development of pericyte coverage of tumor microvessels and aPL-induced tumor cell expression of chemokine ligand 2, a mediator of pericyte recruitment. These findings identify antiphospholipid autoantibodies as a potential patient-specific host factor promoting the transition of indolent tumors to an angiogenic malignant state through a TF-mediated pathogenic mechanism.


Subject(s)
Antibodies, Antiphospholipid/chemistry , Neoplasms/metabolism , Neovascularization, Pathologic , Thromboplastin/metabolism , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Cell Survival , Disease Progression , Endotoxins/chemistry , Female , Gene Expression Regulation , Humans , Immunoglobulin G/chemistry , Mice , Mice, Inbred C57BL , Mice, Nude , Microscopy, Fluorescence , Neoplasm Transplantation
9.
World J Gastroenterol ; 20(30): 10279-87, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25132744

ABSTRACT

Accumulated evidences have demonstrated that signal transducer and activator of transcription 3 (STAT3) is a critical link between inflammation and cancer. Multiple studies have indicated that persistent activation of STAT3 in epithelial/tumor cells in inflammation-associated colorectal cancer (CRC) is associated with sphingosine-1-phosphate (S1P) receptor signaling. In inflammatory response whereby interleukin (IL)-6 production is abundant, STAT3-mediated pathways were found to promote the activation of sphingosine kinases (SphK1 and SphK2) leading to the production of S1P. Reciprocally, S1P encourages the activation of STAT3 through a positive autocrine-loop signaling. The crosstalk between IL-6, STAT3 and sphingolipid regulated pathways may play an essential role in tumorigenesis and tumor progression in inflamed intestines. Therapeutics targeting both STAT3 and sphingolipid are therefore likely to contribute novel and more effective therapeutic strategies against inflammation-associated CRC.


Subject(s)
Colorectal Neoplasms/etiology , Inflammation Mediators/metabolism , Inflammation/complications , Lysophospholipids/metabolism , STAT3 Transcription Factor/metabolism , Sphingosine/analogs & derivatives , Animals , Anti-Inflammatory Agents/therapeutic use , Antineoplastic Agents/therapeutic use , Autocrine Communication , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Humans , Inflammation/drug therapy , Inflammation/immunology , Inflammation/metabolism , Interleukin-6/metabolism , Molecular Targeted Therapy , Signal Transduction , Sphingosine/metabolism
10.
Cell Rep ; 6(4): 625-32, 2014 Feb 27.
Article in English | MEDLINE | ID: mdl-24529703

ABSTRACT

B7-H1 (PD-L1) on immune cells plays an important role in T cell coinhibition by binding its receptor PD-1. Here, we show that both human and mouse intestinal epithelium express B7-H1 and that B7-H1-deficient mice are highly susceptible to dextran sodium sulfate (DSS)- or trinitrobenzenesulfonic acid (TNBS)-induced gut injury. B7-H1 deficiency during intestinal inflammation leads to high mortality and morbidity, which are associated with severe pathological manifestations in the colon, including loss of epithelial integrity and overgrowth of commensal bacteria. Results from bone marrow chimeric and knockout mice show that B7-H1 expressed on intestinal parenchyma, but not on hematopoietic cells, controls intestinal inflammation in an adaptive immunity-independent fashion. Finally, we demonstrate that B7-H1 dampened intestinal inflammation by inhibiting tumor necrosis factor α (TNF-α) production and by stimulating interleukin 22 secretion from CD11c(+)CD11b(+) lamina propria cells. Thus, our data uncover a mechanism through which intestinal tissue-expressed B7-H1 functions as an essential ligand for innate immune cells to prevent gut inflammation.


Subject(s)
Colitis/metabolism , Intestinal Mucosa/metabolism , Programmed Cell Death 1 Receptor/metabolism , Animals , Bone Marrow Cells/metabolism , Colitis/chemically induced , Colitis/immunology , Humans , Immunity, Innate , Inflammation/metabolism , Interleukins/metabolism , Intestinal Mucosa/immunology , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/genetics , Sodium Dodecyl Sulfate/toxicity , Trinitrobenzenesulfonic Acid/toxicity , Tumor Necrosis Factor-alpha/metabolism , Interleukin-22
11.
Neoplasia ; 15(9): 998-1008, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24027425

ABSTRACT

Chronic inflammation is an important risk factor for the development of colorectal cancer; however, the mechanism of tumorigenesis especially tumor progression to malignancy in the inflamed colon is still unclear. Our study shows that epithelial signal transducer and activator of transcription 3 (STAT3), persistently activated in inflamed colon, is not required for inflammation-induced epithelial overproliferation and the development of early-stage tumors; however, it is essential for tumor progression to advanced malignancy. We found that one of the mechanisms that epithelial STAT3 regulates in tumor progression might be to modify leukocytic infiltration in the large intestine. Activation of epithelial STAT3 promotes the infiltration of the CD8+ lymphocyte population but inhibits the recruitment of regulatory T (Treg) lymphocytes. The loss of Stat3 in epithelial cells promoted the expression of cytokines/chemokines including CCL19, CCL28, and RANTES, which are known to be able to recruit Treg lymphocytes. Linked to these changes was the pathway mediated by sphingosine 1-phosphate receptor 1 and sphingosine 1-phosphate kinases, which is activated in colonic epithelial cells in inflamed colon with functional STAT3 but not in epithelial cells deleted of STAT3. Our data suggest that epithelial STAT3 plays a critical role in inflammation-induced tumor progression through regulation of leukocytic recruitment especially the infiltration of Treg cells in the large intestine.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Colitis, Ulcerative/metabolism , Colorectal Neoplasms/metabolism , STAT3 Transcription Factor/metabolism , T-Lymphocytes, Regulatory/immunology , Adenocarcinoma/metabolism , Aged , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Movement/immunology , Cell Proliferation , Chemokine CCL19/biosynthesis , Chemokine CCL5/biosynthesis , Chemokines, CC/biosynthesis , Colon/immunology , Colon/pathology , Epithelial Cells/metabolism , Humans , Inflammation/immunology , Intestinal Mucosa/metabolism , Leukocytes/metabolism , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Receptors, Lysosphingolipid/metabolism , STAT3 Transcription Factor/genetics
12.
mBio ; 3(2)2012.
Article in English | MEDLINE | ID: mdl-22415004

ABSTRACT

UNLABELLED: Annexin A5 (AnxA5) has a high affinity for phosphatidylserine. The protein is widely used to detect apoptotic cells because phosphatidylserine, a phospholipid that is normally present in the inner leaflets of cytoplasmic membranes, becomes translocated to the outer leaflets during programmed cell death. Here we report the novel observation that AnxA5 binds to Gram-negative bacteria via the lipid A domain of lipopolysaccharide (LPS). Binding of AnxA5 to bacteria was measured quantitatively, confirmed by fluorescence microscopy, and found to be inhibited by antibodies against lipid A. AnxA5 also bound to purified dot-blotted LPS and lipid A. Through ellipsometry, we found that the binding of AnxA5 to purified LPS was calcium dependent and rapid and showed a high affinity-characteristics similar to those of AnxA5 binding to phosphatidylserine. Initial functional studies indicated that AnxA5 can affect LPS activities. AnxA5 inhibited LPS-mediated gelation in the Limulus amebocyte lysate assay. Incubation of LPS with the protein reduced the quantity of tumor necrosis factor alpha (TNF-α) released by cultured monocytes compared to that released upon incubation with LPS alone. Initial in vivo experiments indicated that injection of mice with LPS preincubated with AnxA5 produced serum TNF-α levels lower than those seen after injection of LPS alone. These data demonstrate that AnxA5 binds to LPS and open paths to investigation of the potential biological and therapeutic implications of this interaction. IMPORTANCE: AnxA5 is highly expressed in cells that have a barrier function-including, among others, vascular endothelium, placental trophoblasts, and epithelial cells lining bile ducts, renal tubules, mammary ducts, and nasal epithelium. The protein has been well characterized for its binding to phospholipid bilayers that contain phosphatidylserine. This report of a previously unrecognized activity of AnxA5 opens the door to investigation of the possibility that this binding may have biological and therapeutic ramifications. In view of the tissue expression of the protein, the present results suggest the possibility that AnxA5 plays a role in modulating the host defense against lipopolysaccharide at these anatomic sites, where cells may interface with microorganisms. These results also raise the intriguing possibility that AnxA5 or analogous proteins or peptides could provide novel approaches to addressing the difficult clinical problem of Gram-negative sepsis.


Subject(s)
Annexin A5/metabolism , Bacteria/metabolism , Endotoxins/antagonists & inhibitors , Endotoxins/metabolism , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/metabolism , Animals , Calcium/metabolism , Cell Line , Endotoxins/toxicity , Female , Kinetics , Limulus Test , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Monocytes/drug effects , Monocytes/immunology , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/metabolism
13.
Proc Natl Acad Sci U S A ; 108(25): 10272-7, 2011 Jun 21.
Article in English | MEDLINE | ID: mdl-21652773

ABSTRACT

Nutritional and genetic risk factors for intestinal tumors are additive on mouse tumor phenotype, establishing that diet and genetic factors impact risk by distinct combinatorial mechanisms. In a mouse model of dietary-induced sporadic small and large intestinal cancer in WT mice in which tumor etiology, lag, incidence, and frequency reflect >90% of intestinal cancer in Western societies, dietary-induced risk altered gene expression profiles predominantly in villus cells of the histologically normal mucosa, in contrast to targeting of crypt cells by inheritance of an Apc(1638N) allele or homozygous inactivation of p21(Waf1/cip1), and profiles induced by each risk factor were distinct at the gene or functional group level. The dietary-induced changes in villus cells encompassed ectopic expression of Paneth cell markers (a lineage normally confined to the bottom of small intestinal crypts), elevated expression of the Wnt receptor Fzd5 and of EphB2 (genes necessary for Paneth cell differentiation and localization to the crypt bottom), and increased Wnt signaling in villus cells. Ectopic elevation of these markers was also present in the colon crypts, which are also sites of sporadic tumors in the nutritional model. Elevating dietary vitamin D(3) and calcium, which prevents tumor development, abrogated these changes in the villus and colon cells. Thus, common intestinal cancer driven by diet involves mechanisms of tumor development distinct from those mechanisms that cause tumors induced by the rare inheritance of a mutant adenomatous polyposis coli (Apc) allele. This is fundamental for understanding how common sporadic tumors arise and in evaluating relative risk in the population.


Subject(s)
Biomarkers/metabolism , Colon , Colonic Neoplasms/etiology , Diet/adverse effects , Intestinal Mucosa , Intestinal Neoplasms/etiology , Paneth Cells/metabolism , Animals , Cell Transformation, Neoplastic , Colon/cytology , Colon/physiology , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Disease Models, Animal , Gene Expression , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/physiology , Intestinal Neoplasms/genetics , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Mice , Mice, Inbred C57BL , Paneth Cells/cytology , Random Allocation , Risk Factors
15.
Curr HIV Res ; 8(5): 396-404, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20426757

ABSTRACT

Antiretroviral drugs are ineffective at treating viral infection in the brain because they cannot freely diffuse across the blood-brain barrier (BBB). Therefore, HIV-1 viral replication persists in the central nervous system (CNS) and continues to augment the neuropathogenesis process. Nanotechnology can play a pivotal role in HIV-1 therapeutics as it can increase drug solubility, enhance systemic bioavailability, and at the same time offer multifunctionality. Moreover, following conjugation with transferrin (Tf), these drug-loaded nanoformulations can permeate across biological barriers such as the blood brain barrier (BBB) via a receptor mediated transport mechanism. In the current study, we have stably incorporated the antiviral drug, Saquinavir, within Tf-conjugated quantum rods (QRs), which are novel nanoparticles with unique optical properties. We have evaluated the transversing ability of the QR-Tf-Saquinavir nanoformulation across an in vitro model of BBB. In addition, we have analyzed the subsequent antiviral efficacy of this targeted nanoformulation in HIV-1 infected peripheral blood mononuclear cells (PBMCs), which are cultured on the basolateral end of the in vitro BBB model. Our results show a significant uptake of QR-Tf-Saquinavir by brain microvascular endothelial cells (BMVECs), which constitute the BBB. In addition, we observed a significant enhancement in the transversing capability of QR-Tf-Saquinavir across the BBB, along with a marked decrease in HIV-1 viral replication in the PBMCs. These observations indicate that drug-loaded nanoparticles can deliver therapeutics across the BBB. These results highlight the potential of this nanoformulation in the treatment of Neuro-AIDS and other neurological disorders.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Blood-Brain Barrier , Drug Carriers/pharmacokinetics , Nanotubes/chemistry , Saquinavir/pharmacokinetics , Transferrin/pharmacokinetics , Anti-HIV Agents/chemistry , Cell Culture Techniques/methods , Cells, Cultured , Drug Carriers/chemistry , Endothelial Cells/metabolism , HIV-1/drug effects , Humans , Leukocytes, Mononuclear/virology , Saquinavir/chemistry , Transferrin/chemistry , Viral Load
16.
Am J Pathol ; 176(2): 952-67, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20042677

ABSTRACT

Inflammatory bowel disease (IBD) is a high-risk condition for human colorectal cancer. However, our mechanistic understanding of the link between inflammation and tumorigenesis in the colon is limited. Here we established a novel mouse model of colitis-associated cancer by genetically inactivating signal transducer and activator of transcription 3 (Stat3) in macrophages, with partial deletion in other myeloid and lymphoid cells. Inflammation developed in the colon of mutant mice spontaneously, and tumor lesions, including invasive carcinoma, arose in the inflamed region of the intestine with a frequency similar to that observed in human IBD patients. The development of both inflammation and tumors in the mutant mice required the presence of microflora. Indeed, inflammation was associated with disruption of colonic homeostasis, fulminant epithelial/tumor cell proliferation, and activation of the mammalian target of rapamycin (mTOR)-Stat3 pathway in epithelial and tumor cells. The activation of this pathway was essential for both the excess proliferation of epithelial/tumor cells and the disruption of colonic homeostasis in the mutant mice. Notably, a similar abnormal up-regulation of mTOR-Stat3 signaling was consistently observed in the colonic epithelial cells of human IBD patients with active disease. These studies demonstrate a novel mouse model of IBD-colorectal cancer progression in which disrupted immune regulation, mTOR-Stat3 signaling, and epithelial hyperproliferation are integrated and simultaneously linked to the development of malignancy.


Subject(s)
Cell Proliferation , Cell Transformation, Neoplastic/genetics , Disease Models, Animal , Inflammation/genetics , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/pathology , Intracellular Signaling Peptides and Proteins/physiology , Mice , Protein Serine-Threonine Kinases/physiology , Animals , Carcinoma/etiology , Carcinoma/genetics , Carcinoma/pathology , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/pathology , Colitis/complications , Colitis/genetics , Colitis/pathology , Colon/metabolism , Colon/pathology , Colorectal Neoplasms/etiology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Disease Progression , Humans , Inflammation/complications , Inflammation/pathology , Inflammatory Bowel Diseases/complications , Inflammatory Bowel Diseases/genetics , Intestinal Mucosa/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Mice, Inbred C57BL , Mice, Transgenic , Protein Serine-Threonine Kinases/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/physiology , Signal Transduction/genetics , Signal Transduction/physiology , TOR Serine-Threonine Kinases
17.
J Nutr ; 139(11): 2072-8, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19759248

ABSTRACT

A Western-style diet (WD), defined by high-fat, low-calcium, and vitamin D content, is associated with increased risk of human colorectal cancer. Understanding molecular mechanisms altered by the WD is crucial to develop preventive and therapeutic strategies. Effects of a WD on the colonic transcriptome of C57Bl/6J mice, a model for sporadic colon cancer, were studied at endpoints before tumors occur. To assess whether a WD induces inflammatory changes, expression profiles of a broad spectrum of inflammatory proteins were performed and numbers of lamina propria macrophages were determined with semiquantitative morphometry. Transcriptome changes were translated into molecular interaction network maps and pathways. Pathways related to oxidative stress response; lipid, glutathione, and xenobiotic metabolism; and the immune response were perturbed by the WD. Several nuclear factor-erythroid 2-related factor 2- and aryl hydrocarbon receptor-dependent genes, including those coding for enzymes involved in phase 1 and 2 drug metabolism and oxidative stress responses, were induced. Oxidative stress was demonstrated by measurements of endogenous colonic redox-sensitive compound concentrations. Perturbations in immune response-related pathways, expression of inflammatory proteins, and increased numbers of lamina propria macrophages showed that the WD significantly alters the local colonic immune response. Collectively, these data suggest that consumption of a WD interferes with networks of related biological response pathways involving colonic lipid metabolism, oxidative stress, and the immune response. These new findings impact our understanding of links between consumption of WD and colon carcinogenesis, providing additional information for developing preventive means for decreasing colorectal cancer risk.


Subject(s)
Colonic Neoplasms/etiology , Diet/adverse effects , Homeostasis/drug effects , Immunity/drug effects , Oxidative Stress/physiology , Animals , Colorectal Neoplasms/epidemiology , Colorectal Neoplasms/etiology , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , RNA/genetics , RNA/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic/drug effects , Weight Gain
18.
Carcinogenesis ; 30(6): 1016-23, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19359593

ABSTRACT

Curcumin exhibits anti-inflammatory and antitumor activity and is being tested in clinical trials as a chemopreventive agent for colon cancer. Curcumin's chemopreventive activity was tested in a transgenic mouse model of lung cancer that expresses the human Ki-ras(G12C) allele in a doxycycline (DOX) inducible and lung-specific manner. The effects of curcumin were compared with the lung tumor promoter, butylated hydroxytoluene (BHT), and the lung cancer chemopreventive agent, sulindac. Treatment of DOX-induced mice with dietary curcumin increased tumor multiplicity (36.3 +/- 0.9 versus 24.3 +/- 0.2) and progression to later stage lesions, results which were similar to animals that were co-treated with DOX/BHT. Microscopic examination showed that the percentage of lung lesions that were adenomas and adenocarcinomas increased to 66% in DOX/BHT, 66% in DOX/curcumin and 49% in DOX/BHT/curcumin-treated groups relative to DOX only treated mice (19%). Immunohistochemical analysis also showed increased evidence of inflammation in DOX/BHT, DOX/curcumin and DOX/BHT/curcumin mice relative to DOX only treated mice. In contrast, co-treatment of DOX/BHT mice with 200 p.p.m. [DOSAGE ERROR CORRECTED] of sulindac inhibited the progression of lung lesions and reduced the inflammation. Lung tissue from DOX/curcumin-treated mice demonstrated a significant increase (33%; P = 0.01) in oxidative damage, as assessed by the levels of carbonyl protein formation, relative to DOX-treated control mice after 1 week on the curcumin diet. These results suggest that curcumin may exhibit organ-specific effects to enhance reactive oxygen species formation in the damaged lung epithelium of smokers and ex-smokers. Ongoing clinical trials thus may need to exclude smokers and ex-smokers in chemopreventive trials of curcumin.


Subject(s)
Anticarcinogenic Agents/pharmacology , Cell Transformation, Neoplastic/drug effects , Curcumin/pharmacology , Lung Neoplasms/drug therapy , Animals , Anticarcinogenic Agents/adverse effects , Butylated Hydroxytoluene/toxicity , Curcumin/adverse effects , Doxycycline/pharmacology , Genes, ras , Lung Neoplasms/chemically induced , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Transgenic , Organ Specificity , Reactive Oxygen Species/metabolism , Sulindac/pharmacology
19.
Cancer Res ; 67(11): 5064-6, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17545580

ABSTRACT

The development of a supportive vasculature is essential for tumor progression. In a mouse model of breast cancer, we found that tumor-associated macrophages that are recruited to the tumor just before malignant conversion are essential for the angiogenic switch. These findings establish a causal linkage to explain well-documented clinical correlations between macrophages, microvessel density, and poor prognosis in breast tumors.


Subject(s)
Cell Transformation, Neoplastic/pathology , Macrophages/pathology , Mammary Neoplasms, Experimental/blood supply , Animals , Cell Transformation, Neoplastic/immunology , Disease Models, Animal , Macrophages/immunology , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/pathology
20.
Cancer Res ; 67(6): 2649-56, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17363585

ABSTRACT

Although the presence of macrophages in tumors has been correlated with poor prognosis, until now there was no direct observation of how macrophages are involved in hematogenous metastasis. In this study, we use multiphoton microscopy to show, for the first time, that tumor cell intravasation occurs in association with perivascular macrophages in mammary tumors. Furthermore, we show that perivascular macrophages of the mammary tumor are associated with tumor cell intravasation in the absence of local angiogenesis. These results show that the interaction between macrophages and tumor cells lying in close proximity defines a microenvironment that is directly involved in the intravasation of cancer cells in mammary tumors.


Subject(s)
Blood Vessels/pathology , Cell Communication/physiology , Macrophages/pathology , Mammary Neoplasms, Experimental/pathology , Animals , Cell Movement/physiology , Endothelium, Vascular/pathology , Female , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Mice , Microscopy, Fluorescence, Multiphoton , Neoplastic Cells, Circulating/pathology
SELECTION OF CITATIONS
SEARCH DETAIL