Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Howard Li is active.

Publication


Featured researches published by Howard Li.


Molecular Pharmacology | 2007

Antitumorigenic Effects of Peroxisome Proliferator-Activated Receptor-γ in Non-Small-Cell Lung Cancer Cells Are Mediated by Suppression of Cyclooxygenase-2 via Inhibition of Nuclear Factor-κB

Yvette Bren-Mattison; Amy M. Meyer; Vicki Van Putten; Howard Li; Katherine Kuhn; Robert Stearman; Mary C.M. Weiser-Evans; Robert A. Winn; Lynn E. Heasley; Raphael A. Nemenoff

Pharmacological activators of peroxisome proliferator-activated receptor-γ (PPARγ) inhibit growth of non-small-cell lung cancer (NSCLC) cell lines in vitro and in xenograft models. Because these agents engage off-target pathways, we have assessed the effects of PPARγ by overexpressing the protein in NSCLC cells. We reported previously that increased PPARγ inhibits transformed growth and invasiveness and promotes epithelial differentiation in a panel of NSCLC expressing oncogenic K-Ras. These cells express high levels of cyclooxygenase-2 (COX-2) and produce high levels of prostaglandin E2 (PGE2). The goal of these studies was to identify the molecular mechanisms whereby PPARγ inhibits tumorigenesis. Increased PPARγ inhibited expression of COX-2 protein and promoter activity, resulting in decreased PGE2 production. Suppression of COX-2 was mediated through increased activity of the tumor suppressor phosphatase and tensin homolog, leading to decreased levels of phospho-Akt and inhibition of nuclear factor-κB activity. Pharmacological inhibition of PGE2 production mimicked the effects of PPARγ on epithelial differentiation in three-dimensional culture, and exogenous PGE2 reversed the effects of increased PPARγ activity. Transgenic mice overexpressing PPARγ under the control of the surfactant protein C promoter had reduced expression of COX-2 in type II cells and were protected against developing lung tumors in a chemical carcinogenesis model. These data indicate that high levels of PGE2 as a result of elevated COX-2 expression are critical for promoting lung tumorigenesis and that the antitumorigenic effects of PPARγ are mediated in part through blocking this pathway.


Clinical Cancer Research | 2012

Loss of Transforming Growth Factor Beta Type II Receptor Increases Aggressive Tumor Behavior and Reduces Survival in Lung Adenocarcinoma and Squamous Cell Carcinoma

Stephen P. Malkoski; Sarah M. Haeger; Timothy G. Cleaver; Karen J. Rodriguez; Howard Li; Shi-Long Lu; William J. Feser; Anna E. Barón; Daniel T. Merrick; Jessyka G. Lighthall; Hideaki Ijichi; Wilbur A. Franklin; Xiao-Jing Wang

Purpose: Lung adenocarcinoma and lung squamous cell carcinoma (SCC) are the most common non–small cell lung cancer (NSCLC) subtypes. This study was designed to determine whether reduced expression of TGFβ type II receptor (TGFβRII) promotes lung adenocarcinoma and SCC carcinogenesis. Experimental Design: We examined TGFβRII expression at the protein and mRNA levels in human NSCLC samples and assessed the relationship between TGFβRII expression and clinicopathologic parameters. To determine whether sporadic TGFβRII deletion in airway epithelial cells induces NSCLC formation, we targeted TGFβRII deletion alone and in combination with oncogenic KrasG12D to murine airways using a keratin 5 (K5) promoter and inducible Cre recombinase. Results: Reduced TGFβRII expression in human NSCLC is associated with male gender, smoking, SCC histology, reduced differentiation, increased tumor stage, increased nodal metastasis, and reduced survival. Homozygous or heterozygous TGFβRII deletion in mouse airway epithelia increases the size and number of KrasG12D-initiated adenocarcinoma and SCC. TGFβRII deletion increases proliferation, local inflammation, and TGFβ ligand elaboration; TGFβRII knockdown in airway epithelial cells increases migration and invasion. Conclusions: Reduced TGFβRII expression in human NSCLC is associated with more aggressive tumor behavior and inflammation that is, at least partially, mediated by increased TGFβ1 expression. TGFβRII deletion in mouse airway epithelial cells promotes adenocarcinoma and SCC formation, indicating that TGFβRII loss plays a causal role in lung carcinogenesis. That TGFβRII shows haploid insufficiency suggests that a 50% TGFβRII protein reduction would negatively impact lung cancer prognosis. Clin Cancer Res; 18(8); 2173–83. ©2012 AACR.


PLOS ONE | 2011

Activation of PPARγ in myeloid cells promotes lung cancer progression and metastasis.

Howard Li; Amber L. Sorenson; Joanna M. Poczobutt; Jay Amin; Teresa Joyal; Timothy M. Sullivan; Joseph T. Crossno; Mary C.M. Weiser-Evans; Raphael A. Nemenoff

Activation of peroxisome proliferator-activated receptor-γ (PPARγ) inhibits growth of cancer cells including non-small cell lung cancer (NSCLC). Clinically, use of thiazolidinediones, which are pharmacological activators of PPARγ is associated with a lower risk of developing lung cancer. However, the role of this pathway in lung cancer metastasis has not been examined well. The systemic effect of pioglitazone was examined in two models of lung cancer metastasis in immune-competent mice. In an orthotopic model, murine lung cancer cells implanted into the lungs of syngeneic mice metastasized to the liver and brain. As a second model, cancer cells injected subcutaneously metastasized to the lung. In both models systemic administration of pioglitazone increased the rate of metastasis. Examination of tissues from the orthotopic model demonstrated increased numbers of arginase I-positive macrophages in tumors from pioglitazone-treated animals. In co-culture experiments of cancer cells with bone marrow-derived macrophages, pioglitazone promoted arginase I expression in macrophages and this was dependent on the expression of PPARγ in the macrophages. To assess the contribution of PPARγ in macrophages to cancer progression, experiments were performed in bone marrow-transplanted animals receiving bone marrow from Lys-M-Cre+/PPARγflox/flox mice, in which PPARγ is deleted specifically in myeloid cells (PPARγ-Macneg), or control PPARγflox/flox mice. In both models, mice receiving PPARγ-Macneg bone marrow had a marked decrease in secondary tumors which was not significantly altered by treatment with pioglitazone. This was associated with decreased numbers of arginase I-positive cells in the lung. These data support a model in which activation of PPARγ may have opposing effects on tumor progression, with anti-tumorigenic effects on cancer cells, but pro-tumorigenic effects on cells of the microenvironment, specifically myeloid cells.


Molecular Biology of the Cell | 2008

Oncogenic K-Ras Regulates Proliferation and Cell Junctions in Lung Epithelial Cells through Induction of Cyclooxygenase-2 and Activation of Metalloproteinase-9

Xue Qing Wang; Howard Li; Vicki Van Putten; Robert A. Winn; Lynn E. Heasley; Raphael A. Nemenoff

Expression of oncogenic K-Ras is frequently observed in non-small-cell lung cancer. However, oncogenic K-Ras is not sufficient to transform lung epithelial cells and requires collaborating signals that have not been defined. To examine the biological effects of K-Ras in nontransformed lung epithelial cells, stable transfectants were generated in RL-65 cells, a spontaneously immortalized lung epithelial cell line. Expression of K-Ras resulted in extracellular signal-regulated kinase (ERK) activation, which mediated induction of cyclooxygenase (COX)-2 and increased prostaglandin E(2) production. Epithelial cells expressing oncogenic K-Ras showed increased proliferation in two- and three-dimensional tissue culture and delayed formation of hollow acinar structures in three-dimensional matrigel cultures. These affects were mediated through COX-2-dependent activation of beta-catenin signaling and inhibition of apoptosis. ERK activation also led to induction of metalloproteinase (MMP)-9 and cleavage of E-cadherin at two specific sites. This resulted in partial disruption of adherens junctions as determined by decreased transepithelial resistance (TER), and disruption of E-cadherin/beta-catenin interactions. An MMP-9 inhibitor reversed the decrease in TER and inhibited beta-catenin signaling. These data indicate that although expression of oncogenic K-Ras does not transform lung epithelial cells, it alters the phenotype of the cells by increasing proliferation and decreasing cell-cell contacts characteristic of epithelial cells.


Journal of Immunology | 2016

Deletion of 5-Lipoxygenase in the Tumor Microenvironment Promotes Lung Cancer Progression and Metastasis through Regulating T Cell Recruitment

Joanna M. Poczobutt; Teresa T. Nguyen; Dwight Hanson; Howard Li; Trisha R. Sippel; Mary C.M. Weiser-Evans; Miguel A. Gijón; Robert C. Murphy; Raphael A. Nemenoff

Eicosanoids, including PGs, produced by cyclooxygenases (COX), and leukotrienes, produced by 5-lipoxygenase (5-LO) have been implicated in cancer progression. These molecules are produced by both cancer cells and the tumor microenvironment (TME). We previously reported that both COX and 5-LO metabolites increase during progression in an orthotopic immunocompetent model of lung cancer. Although PGs in the TME have been well studied, less is known regarding 5-LO products produced by the TME. We examined the role of 5-LO in the TME using a model in which Lewis lung carcinoma cells are directly implanted into the lungs of syngeneic WT mice or mice globally deficient in 5-LO (5-LO-KO). Unexpectedly, primary tumor volume and liver metastases were increased in 5-LO-KO mice. This was associated with an ablation of leukotriene (LT) production, consistent with production mainly mediated by the microenvironment. Increased tumor progression was partially reproduced in global LTC4 synthase KO or mice transplanted with LTA4 hydrolase-deficient bone marrow. Tumor-bearing lungs of 5-LO-KO had decreased numbers of CD4 and CD8 T cells compared with WT controls, as well as fewer dendritic cells. This was associated with lower levels of CCL20 and CXL9, which have been implicated in dendritic and T cell recruitment. Depletion of CD8 cells increased tumor growth and eliminated the differences between WT and 5-LO mice. These data reveal an antitumorigenic role for 5-LO products in the microenvironment during lung cancer progression through regulation of T cells and suggest that caution should be used in targeting this pathway in lung cancer.


PLOS ONE | 2013

Eicosanoid profiling in an orthotopic model of lung cancer progression by mass spectrometry demonstrates selective production of leukotrienes by inflammatory cells of the microenvironment.

Joanna M. Poczobutt; Miguel A. Gijón; Jay Amin; Dwight Hanson; Howard Li; Deandra Walker; Mary C.M. Weiser-Evans; Xian Lu; Robert C. Murphy; Raphael A. Nemenoff

Eicosanoids are bioactive lipid mediators derived from arachidonic acid1 (AA), which is released by cytosolic phospholipase A2 (cPLA2). AA is metabolized through three major pathways, cyclooxygenase (COX), lipoxygenase (LO) and cytochrome P450, to produce a family of eicosanoids, which individually have been shown to have pro- or anti-tumorigenic activities in cancer. However, cancer progression likely depends on complex changes in multiple eicosanoids produced by cancer cells and by tumor microenvironment and a systematic examination of the spectrum of eicosanoids in cancer has not been performed. We used liquid chromatography coupled with tandem mass spectrometry (LC/MS/MS) to quantitate eicosanoids produced during lung tumor progression in an orthotopic immunocompetent mouse model of lung cancer, in which Lewis lung carcinoma (LLC) cells are injected into lungs of syngeneic mice. The presence of tumor increased products of both the cyclooxygenase and the lipoxygenase pathways in a time-dependent fashion. Comparing tumors grown in cPLA2 knockout vs wild-type mice, we demonstrated that prostaglandins (PGE2, PGD2 and PGF2a) were produced by both cancer cells and the tumor microenvironment (TME), but leukotriene (LTB4, LTC4, LTD4, LTE4) production required cPLA2 expression in the TME. Using flow cytometry, we recovered tumor-associated neutrophils and 2 types of tumor-associated macrophages from tumor-bearing lungs and we defined their distinct eicosanoid profiles by LC/MS/MS. The combination of flow cytometry and LC/MS/MS unravels the complexity of eicosanoid production in lung cancer and provides a rationale to develop therapeutic strategies that target select cell populations to inhibit specific classes of eicosanoids.


Cancer Research | 2014

Homeoprotein Six2 promotes breast cancer metastasis via transcriptional and epigenetic control of E-cadherin expression

Chu-An Wang; David J. Drasin; Catherine Pham; Paul Jedlicka; Vadym Zaberezhnyy; Michelle Guney; Howard Li; Raphael A. Nemenoff; James C. Costello; Aik Choon Tan; Heide L. Ford

Misexpression of developmental transcription factors occurs often in human cancers, where embryonic programs may be reinstated in a context that promotes or sustains malignant development. In this study, we report the involvement of the kidney development transcription factor Six2 in the metastatic progression of human breast cancer. We found that Six2 promoted breast cancer metastasis by a novel mechanism involving both transcriptional and epigenetic regulation of E-cadherin. Downregulation of E-cadherin by Six2 was necessary for its ability to increase soft agar growth and in vivo metastasis in an immunocompetent mouse model of breast cancer. Mechanistic investigations showed that Six2 represses E-cadherin expression by upregulating Zeb2, in part, through a microRNA-mediated mechanism and by stimulating promoter methylation of the E-cadherin gene (Cdh1). Clinically, SIX2 expression correlated inversely with CDH1 expression in human breast cancer specimens, corroborating the disease relevance of their interaction. Our findings establish Six2 as a regulator of metastasis in human breast cancers and demonstrate an epigenetic function for SIX family transcription factors in metastatic progression through the regulation of E-cadherin.


Journal of Immunology | 2016

Expression Profiling of Macrophages Reveals Multiple Populations with Distinct Biological Roles in an Immunocompetent Orthotopic Model of Lung Cancer.

Joanna M. Poczobutt; Subhajyoti De; Vinod Kumar Yadav; Teresa T. Nguyen; Howard Li; Trisha R. Sippel; Mary C.M. Weiser-Evans; Raphael A. Nemenoff

Macrophages represent an important component of the tumor microenvironment and play a complex role in cancer progression. These cells are characterized by a high degree of plasticity, and they alter their phenotype in response to local environmental cues. Whereas the M1/M2 classification of macrophages has been widely used, the complexity of macrophage phenotypes has not been well studied, particularly in lung cancer. In this study we employed an orthotopic immunocompetent model of lung adenocarcinoma in which murine lung cancer cells are directly implanted into the left lobe of syngeneic mice. Using multimarker flow cytometry, we defined and recovered several distinct populations of monocytes/macrophages from tumors at different stages of progression. We used RNA-seq transcriptional profiling to define distinct features of each population and determine how they change during tumor progression. We defined an alveolar resident macrophage population that does not change in number and expresses multiple genes related to lipid metabolism and lipid signaling. We also defined a population of tumor-associated macrophages that increase dramatically with tumor and selectively expresses a panel of chemokine genes. A third population, which resembles tumor-associated monocytes, expresses a large number of genes involved in matrix remodeling. By correlating transcriptional profiles with clinically prognostic genes, we show that specific monocyte/macrophage populations are enriched in genes that predict outcomes in lung adenocarcinoma, implicating these subpopulations as critical determinants of patient survival. Our data underscore the complexity of monocytes/macrophages in the tumor microenvironment, and they suggest that distinct populations play specific roles in tumor progression.


Journal of Molecular Medicine | 2016

CD4 T cell knockout does not protect against kidney injury and worsens cancer

Kameswaran Ravichandran; Qian Wang; Abdullah Ozkok; Alkesh Jani; Howard Li; Zhibin He; Danica Galešić Ljubanović; Mary C.M. Weiser-Evans; Raphael A. Nemenoff; Charles L. Edelstein

Most previous studies of cisplatin-induced acute kidney injury (AKI) have been in models of acute, high-dose cisplatin administration that leads to mortality in non-tumor-bearing mice. The aim of the study was to determine whether CD4 T cell knockout protects against AKI and cancer in a clinically relevant model of low-dose cisplatin-induced AKI in mice with cancer. Kidney function, serum neutrophil gelatinase-associated lipocalin (NGAL), acute tubular necrosis (ATN), and tubular apoptosis score were the same in wild-type and CD4 −/− mice with AKI. The lack of protection against AKI in CD4 −/− mice was associated with an increase in extracellular signal-regulated kinase (ERK), p38, CXCL1, and TNF-α, mediators of AKI and fibrosis, in both cisplatin-treated CD4 −/− mice and wild-type mice. The lack of protection was independent of the presence of cancer or not. Tumor size was double, and cisplatin had an impaired therapeutic effect on the tumors in CD4 −/− vs. wild-type mice. Mice depleted of CD4 T cells using the GK1.5 antibody were not protected against AKI and had larger tumors and lesser response to cisplatin. In summary, in a clinically relevant model of cisplatin-induced AKI in mice with cancer, (1) CD4 −/− mice were not protected against AKI; (2) ERK, p38, CXCL1, and TNF-α, known mediators of AKI, and interstitial fibrosis were increased in CD4 −/− kidneys; and (3) CD4 −/− mice had faster tumor growth and an impaired therapeutic effect of cisplatin on the tumors. The data warns against the use of CD4 T cell inhibition to attenuate cisplatin-induced AKI in patients with cancer.Key messageA clinically relevant low-dose cisplatin model of AKI in mice with cancer was used.CD4 −/− mice were not functionally or histologically protected against AKI.CD4 −/− mice had faster tumor growth.CD4 −/− mice had an impaired therapeutic effect of cisplatin on the tumors.Mice depleted of CD4 T cells were not protected against AKI and had larger tumors.


Chest | 2009

Dyspnea in a 43-Year-Old Woman With Polycystic Kidney Disease

Howard Li; Gregory P. Cosgrove; Jeffrey J. Swigris

A 43-year-old Vietnamese-American woman was referred for evaluation of dyspnea. Six months prior to presentation, left-sided chest pain radiating to her shoulder had developed, and she also noted nonproductive cough and decreased exercise tolerance. These symptoms resolved spontaneously after 10 days, and she returned to her usual state of good health. Within a few weeks began the insidious onset of progressive exertion-related dyspnea. She denied fevers, night sweats, weight loss, cough, sputum production, hemoptysis, wheezing, or recurrent chest pain. She was seen by her primary care physician who started her on loratadine, 10 mg/d, and albuterol by metered-dose inhaler. When her symptoms failed to improve, she was referred to our institution. Her medical history was significant for hypertension, hyperlipidemia, and polycystic kidney disease. Her medications at the time of presentation to our center included hydrochlorothiazide, simvastatin, lansoprazole, and loratadine. She was a lifetime nonsmoker, denied recent travel, and had no risk factors for tuberculosis. There was no known family history of polycystic kidney or parenchymal lung disease. On physical examination, vital signs were normal and pulse-derived oxygen saturation was 98% on room air. Lung auscultation revealed diminished breath sounds bilaterally, but without crackles, wheeze, or rhonchi. She was not clubbed, nor did she have peripheral edema, rash, or skin lesions. Neurologic examination was unremarkable. Results for CBC count, chemistry panel, liver tests, and coagulation studies were normal. Room air, resting blood gas showed pH 7.45, PaCO2 of 33 mm Hg, and PaO2 of 71 mm Hg. Urinalysis was normal. Spot urine protein/creatinine ratio was 0.1. Spirometry showed the following: FVC, 3.67 L (86% of predicted); FEV1, 2.92 L (85% of predicted); and FEV1/FVC, 0.80. Lung volumes measured by body plethysmograph revealed total gas volume of 4.01 L (123% of predicted), residual volume of 3.25 L (171% of predicted), and total lung capacity of 6.82 L (118% of predicted). Diffusion capacity of the lung for carbon monoxide unadjusted for hemoglobin was 90% of predicted. High-resolution CT of the chest and unenhanced CT scans of the abdomen and pelvis were performed (Fig 1, ​,22). Figure 1 Chest CT scan demonstrating innumerable thin-walled cysts of varying sizes randomly distributed throughout the parenchyma of both lungs. Figure 2 Abdominal CT scan showing massive kidneys with innumerable fluid-filled cysts.

Collaboration


Dive into the Howard Li's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joanna M. Poczobutt

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Trisha R. Sippel

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Amber Sorenson

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Eric T. Clambey

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Jeff Kwak

Anschutz Medical Campus

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Robert A. Winn

University of Colorado Hospital

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge