Network


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

Hotspot


Dive into the research topics where Mohammad W. Khan is active.

Publication


Featured researches published by Mohammad W. Khan.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Regulation of induced colonic inflammation by Lactobacillus acidophilus deficient in lipoteichoic acid

Mansour Mohamadzadeh; Erika A. Pfeiler; Jeffrey B. Brown; Mojgan Zadeh; Matthew Gramarossa; Elizabeth Managlia; Praveen Bere; Bara Sarraj; Mohammad W. Khan; Krishna Chaitanya Pakanati; M. Javeed Ansari; Sarah O'Flaherty; Terrence A. Barrett; Todd R. Klaenhammer

Imbalance in the regulatory immune mechanisms that control intestinal cellular and bacterial homeostasis may lead to induction of the detrimental inflammatory signals characterized in humans as inflammatory bowel disease. Induction of proinflammatory cytokines (i.e., IL-12) induced by dendritic cells (DCs) expressing pattern recognition receptors may skew naive T cells to T helper 1 polarization, which is strongly implicated in mucosal autoimmunity. Recent studies show the ability of probiotic microbes to treat and prevent numerous intestinal disorders, including Clostridium difficile-induced colitis. To study the molecular mechanisms involved in the induction and repression of intestinal inflammation, the phosphoglycerol transferase gene that plays a key role in lipoteichoic acid (LTA) biosynthesis in Lactobacillus acidophilus NCFM (NCK56) was deleted. The data show that the L. acidophilus LTA-negative in LTA (NCK2025) not only down-regulated IL-12 and TNFα but also significantly enhanced IL-10 in DCs and controlled the regulation of costimulatory DC functions, resulting in their inability to induce CD4+ T-cell activation. Moreover, treatment of mice with NCK2025 compared with NCK56 significantly mitigated dextran sulfate sodium and CD4+CD45RBhighT cell-induced colitis and effectively ameliorated dextran sulfate sodium-established colitis through a mechanism that involves IL-10 and CD4+FoxP3+ T regulatory cells to dampen exaggerated mucosal inflammation. Directed alteration of cell surface components of L. acidophilus NCFM establishes a potential strategy for the treatment of inflammatory intestinal disorders.


Cancer and Metastasis Reviews | 2011

The significant role of mast cells in cancer

Khashayarsha Khazaie; Nichole R. Blatner; Mohammad W. Khan; Fotini Gounari; Elias Gounaris; Kristen L. Dennis; Andreas Bonertz; Fu Nien Tsai; Matthew J. Strouch; Eric C. Cheon; Joseph D. Phillips; David J. Bentrem

Mast cells (MC) are a bone marrow-derived, long-lived, heterogeneous cellular population that function both as positive and negative regulators of immune responses. They are arguably the most productive chemical factory in the body and influence other cells through both soluble mediators and cell-to-cell interaction. MC are commonly seen in various tumors and have been attributed alternatively with tumor rejection or tumor promotion. Tumor-infiltrating MC are derived both from sentinel and recruited progenitor cells. MC can directly influence tumor cell proliferation and invasion but also help tumors indirectly by organizing its microenvironment and modulating immune responses to tumor cells. Best known for orchestrating inflammation and angiogenesis, the role of MC in shaping adaptive immune responses has become a focus of recent investigations. MC mobilize T cells and antigen-presenting dendritic cells. They function as intermediaries in regulatory T cells (Treg)-induced tolerance but can also modify or reverse Treg-suppressive properties. The central role of MC in the control of innate and adaptive immunity endows them with the ability to tune the nature of host responses to cancer and ultimately influence the outcome of disease and fate of the cancer patient.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Abating colon cancer polyposis by Lactobacillus acidophilus deficient in lipoteichoic acid

Khashayarsha Khazaie; Mojgan Zadeh; Mohammad W. Khan; Praveen Bere; Fotini Gounari; Kirsten Dennis; Nichole R. Blatner; Jennifer L. Owen; Todd R. Klaenhammer; Mansour Mohamadzadeh

An imbalance of commensal bacteria and their gene products underlies mucosal and, in particular, gastrointestinal inflammation and a predisposition to cancer. Lactobacillus species have received considerable attention as examples of beneficial microbiota. We have reported previously that deletion of the phosphoglycerol transferase gene that is responsible for lipoteichoic acid (LTA) biosynthesis in Lactobacillus acidophilus (NCK2025) rendered this bacterium able to significantly protect mice against induced colitis when delivered orally. Here we report that oral treatment with LTA-deficient NCK2025 normalizes innate and adaptive pathogenic immune responses and causes regression of established colonic polyps. This study reveals the proinflammatory role of LTA and the ability of LTA-deficient L. acidophilus to regulate inflammation and protect against colonic polyposis in a unique mouse model.


Science Translational Medicine | 2012

Expression of RORγt Marks a Pathogenic Regulatory T Cell Subset in Human Colon Cancer

Nichole R. Blatner; Mary F. Mulcahy; Kristen L. Dennis; Denise M. Scholtens; David J. Bentrem; Joseph D. Phillips; Soo Ham; Barry P. Sandall; Mohammad W. Khan; David M. Mahvi; Amy L. Halverson; Steven J. Stryker; Anne Marie Boller; Ashima Singal; Rebekka K. Sneed; Bara Sarraj; Mohammed Javeed Ansari; Martin Oft; Yoichiro Iwakura; Liang Zhou; Andreas Bonertz; Fotini Gounari; Khashayarsha Khazaie

Tregs that expand in human colon cancer have proinflammatory properties and contribute to tumor progression. A Treg Melting Pot Some things are not what they seem. Like the allegorical wolf in sheep’s clothing, cell populations that may seem homogeneous may actually contain subsets with different functions. Indeed, such hidden subpopulations may result in contradictory findings in different systems. Blatner et al. now find a subset of regulatory T cells (Tregs) in human colon cancer that may explain disparate clinical outcomes between studies. The authors found preferential expansion in human colon cancer of Tregs that can suppress T cells but are not anti-inflammatory like more classic Tregs. They then looked in a mouse model of hereditary polyposis and found that these cells, which express Foxp3 and RORγt, express the proinflammatory cytokine IL-17 and are directly associated with inflammation and disease progression. The balance between anti-inflammatory Tregs and these “pathogenic” proinflammatory Tregs may play a role in regulating cancer inflammation. Targeting these RORγt+ Tregs may influence disease outcome in colon cancer. The role of regulatory T cells (Tregs) in human colon cancer (CC) remains controversial: high densities of tumor-infiltrating Tregs can correlate with better or worse clinical outcomes depending on the study. In mouse models of cancer, Tregs have been reported to suppress inflammation and protect the host, suppress T cells and protect the tumor, or even have direct cancer-promoting attributes. These different effects may result from the presence of different Treg subsets. We report the preferential expansion of a Treg subset in human CC with potent T cell–suppressive, but compromised anti-inflammatory, properties; these cells are distinguished from Tregs present in healthy donors by their coexpression of Foxp3 and RORγt. Tregs with similar attributes were found to be expanded in mouse models of hereditary polyposis. Indeed, ablation of the RORγt gene in Foxp3+ cells in polyp-prone mice stabilized Treg anti-inflammatory functions, suppressed inflammation, improved polyp-specific immune surveillance, and severely attenuated polyposis. Ablation of interleukin-6 (IL-6), IL-23, IL-17, or tumor necrosis factor–α in polyp-prone mice reduced polyp number but not to the same extent as loss of RORγt. Surprisingly, loss of IL-17A had a dual effect: IL-17A–deficient mice had fewer polyps but continued to have RORγt+ Tregs and developed invasive cancer. Thus, we conclude that RORγt has a central role in determining the balance between protective and pathogenic Tregs in CC and that Treg subtype regulates inflammation, potency of immune surveillance, and severity of disease outcome.


Journal of Biological Chemistry | 2013

Tumor STAT1 Transcription Factor Activity Enhances Breast Tumor Growth and Immune Suppression Mediated by Myeloid-derived Suppressor Cells

Laura M. Hix; John Karavitis; Mohammad W. Khan; Yihui H. Shi; Khashayarsha Khazaie; Ming Zhang

Background: The role of STAT1 in promoting tumor progression is not well understood. Results: STAT1 activity is elevated in human and mouse breast cancers, and STAT1 promotes breast cancer progression. Conclusion: Tumor STAT1 transcription factor activity enhances breast tumor growth and immune suppression mediated by MDSCs. Significance: STAT1 activity in breast cancer cells is responsible for shaping an immunosuppressive tumor microenvironment, and inhibiting STAT1 activity is a promising immune therapeutic approach. Previous studies had implicated the IFN-γ transcription factor signal transducer and activator of transcription 1 (STAT1) as a tumor suppressor. However, accumulating evidence has correlated increased STAT1 activation with increased tumor progression in multiple types of cancer, including breast cancer. Indeed, we present evidence that tumor up-regulation of STAT1 activity in human and mouse mammary tumors correlates with increasing disease progression to invasive carcinoma. A microarray analysis comparing low aggressive TM40D and highly aggressive TM40D-MB mouse mammary carcinoma cells revealed significantly higher STAT1 activity in the TM40D-MB cells. Ectopic overexpression of constitutively active STAT1 in TM40D cells promoted mobilization of myeloid-derived suppressor cells (MDSCs) and inhibition of antitumor T cells, resulting in aggressive tumor growth in tumor-transplanted, immunocompetent mice. Conversely, gene knockdown of STAT1 in the metastatic TM40D-MB cells reversed these events and attenuated tumor progression. Importantly, we demonstrate that in human breast cancer, the presence of tumor STAT1 activity and tumor-recruited CD33+ myeloid cells correlates with increasing disease progression from ductal carcinoma in situ to invasive carcinoma. We conclude that STAT1 activity in breast cancer cells is responsible for shaping an immunosuppressive tumor microenvironment, and inhibiting STAT1 activity is a promising immune therapeutic approach.


Cancer Research | 2011

Mast cell 5-lipoxygenase activity promotes intestinal polyposis in APC Δ468 mice

Eric C. Cheon; Khashayarsha Khazaie; Mohammad W. Khan; Matthew J. Strouch; Seth B. Krantz; Joseph D. Phillips; Nichole R. Blatner; Laura M. Hix; Ming Zhang; Kristen L. Dennis; Mohammed R. Salabat; Michael J. Heiferman; Paul J. Grippo; Hidayatullah G. Munshi; Elias Gounaris; David J. Bentrem

Arachidonic acid metabolism has been implicated in colon carcinogenesis, but the role of hematopoietic 5-lipoxygenase (5LO) that may impact tumor immunity in development of colon cancer has not been explored. Here we show that tissue-specific deletion of the 5LO gene in hematopoietic cells profoundly attenuates polyp development in the APC(Δ468) murine model of colon polyposis. In vitro analyses indicated that mast cells in particular utilized 5LO to limit proliferation of intestinal epithelial cells and to mobilize myeloid-derived suppressor cells (MDSCs). Mice lacking hemapoietic expression of 5LO exhibited reduced recruitment of MDSCs to the spleen, mesenteric lymph nodes, and primary tumor site. 5LO deficiency also reduced the activity in MDSCs of arginase-1, which is thought to be critical for MDSC function. Together, our results establish a pro-tumorigenic role of hematopoietic 5LO in the immune microenvironment and suggest 5LO inhibition as an avenue for future investigation in treatment of colorectal polyposis and cancer.


Science Translational Medicine | 2014

β-Catenin Promotes Colitis and Colon Cancer Through Imprinting of Proinflammatory Properties in T Cells

Shilpa Keerthivasan; Katayoun Aghajani; Marei Dose; Luciana Molinero; Mohammad W. Khan; Vysak Venkateswaran; Christopher R. Weber; Akinola Olumide Emmanuel; Tianjao Sun; David J. Bentrem; Mary F. Mulcahy; Ali Keshavarzian; Elena M. Ramos; Nichole R. Blatner; Khashayarsha Khazaie; Fotini Gounari

Wnt/β-catenin signaling in T cells is activated during polyposis and colon cancer and drives inflammation and tumor growth by promoting expression of TH17-associated genes including RORγt. β-Catenin, Corrupter of the Tregs It is well known that the numbers and types of lymphocytes that infiltrate colon tumors are relevant to the clinical outcome. However, the reasons for this association are complex and not yet fully understood. Here, Keerthivasan and colleagues identify β-catenin as a culprit and show that it directly contributes to inflammation and colon carcinogenesis in patients and mice with underlying colitis. Keerthivasan and coauthors also provide a mechanistic explanation for the observed effects, showing that the expression of β-catenin in T cells induces the expression of T helper 17 (TH17) genes. This activation of TH17 genes converts T cells to a proinflammatory phenotype and impairs Treg development. Mice overexpressing β-catenin all develop colitis and then colitis-induced cancer, even when the β-catenin is overexpressed only in Tregs. Similarly, human patients with colitis-induced or sporadic colon cancer have abnormally high amounts of β-catenin–expressing T cells in their tumors, confirming the relevance of the mouse findings to human disease. The density and type of lymphocytes that infiltrate colon tumors are predictive of the clinical outcome of colon cancer. High densities of T helper 17 (TH17) cells and inflammation predict poor outcome, whereas infiltration by T regulatory cells (Tregs) that naturally suppress inflammation is associated with longer patient survival. However, the role of Tregs in cancer remains controversial. We recently reported that Tregs in colon cancer patients can become proinflammatory and tumor-promoting. These properties were directly linked with their expression of RORγt (retinoic acid–related orphan receptor-γt), the signature transcription factor of TH17 cells. We report that Wnt/β-catenin signaling in T cells promotes expression of RORγt. Expression of β-catenin was elevated in T cells, including Tregs, of patients with colon cancer. Genetically engineered activation of β-catenin in mouse T cells resulted in enhanced chromatin accessibility in the proximity of T cell factor-1 (Tcf-1) binding sites genome-wide, induced expression of TH17 signature genes including RORγt, and promoted TH17-mediated inflammation. Strikingly, the mice had inflammation of small intestine and colon and developed lesions indistinguishable from colitis-induced cancer. Activation of β-catenin only in Tregs was sufficient to produce inflammation and initiate cancer. On the basis of these findings, we conclude that activation of Wnt/β-catenin signaling in effector T cells and/or Tregs is causatively linked with the imprinting of proinflammatory properties and the promotion of colon cancer.


Clinical Cancer Research | 2013

PI3K/AKT Signaling Is Essential for Communication between Tissue-Infiltrating Mast Cells, Macrophages, and Epithelial Cells in Colitis-Induced Cancer

Mohammad W. Khan; Ali Keshavarzian; Elias Gounaris; Joshua E. Melson; Eric C. Cheon; Nichole R. Blatner; Zongmin Eric Chen; Fu-Nien Tsai; Goo Lee; Hyunji Ryu; Terrence A. Barrett; David J. Bentrem; Khashayarsha Khazaie

Purpose: To understand signaling pathways that shape inflamed tissue and predispose to cancer is critical for effective prevention and therapy for chronic inflammatory diseases. We have explored phosphoinositide 3-kinase (PI3K) activity in human inflammatory bowel diseases and mouse colitis models. Experimental Design: We conducted immunostaining of phosphorylated AKT (pAKT) and unbiased high-throughput image acquisition and quantitative analysis of samples of noninflamed normal colon, colitis, dysplasia, and colorectal cancer. Mechanistic insights were gained from ex vivo studies of cell interactions, the piroxicam/IL-10−/− mouse model of progressive colitis, and use of the PI3K inhibitor LY294002. Results: Progressive increase in densities of pAKT-positive tumor-associated macrophages (TAM) and increase in densities of mast cells in the colonic submucosa were noted with colitis and progression to dysplasia and cancer. Mast cells recruited macrophages in ex vivo migration assays, and both mast cells and TAMs promoted invasion of cancer cells. Pretreatment of mast cells with LY294002 blocked recruitment of TAMs. LY294002 inhibited mast cell and TAM-mediated tumor invasion, and in mice, blocked stromal PI3K, colitis, and cancer. Conclusion: The PI3K/AKT pathway is active in cells infiltrating inflamed human colon tissue. This pathway sustains the recruitment of inflammatory cells through a positive feedback loop. The PI3K/AKT pathway is essential for tumor invasion and the malignant features of the piroxicam/IL-10−/− mouse model. LY294002 targets the PI3K pathway and hinders progressive colitis. These findings indicate that colitis and progression to cancer are dependent on stromal PI3K and sensitive to treatment with LY294002. Clin Cancer Res; 19(9); 2342–54. ©2013 AACR.


Inflammatory Bowel Diseases | 2013

Fluorescence endoscopy of cathepsin activity discriminates dysplasia from colitis

Elias Gounaris; John Martin; Yasushige Ishihara; Mohammad W. Khan; Goo Lee; Preetika Sinh; Eric Zongming Chen; Michael Angarone; Ralph Weissleder; Khasharyasha Khazaie; Terrence A. Barrett

Background:Surveillance colonoscopy using random biopsies to detect colitis-associated cancer (CAC) suffers from poor sensitivity. Although chromoendoscopy improves detection, acceptance in the community has been slow. Here, we examine the usefulness of near infrared fluorescence (NIRF) endoscopy to image molecular probes for cathepsin activity in colitis-induced dysplasia. Methods:In patient samples, cathepsin activity was correlated with colitis and dysplasia. In mice, cathepsin activity was detected as fluorescent hydrolysis product of substrate-based probes after injection into Il10−/− colitic mice. Fluorescence colonoscopy and colonic whole-mount imaging were performed before complete sectioning and pathology review of resected colons. Results:Cathepsin activity was 5-fold and 8-fold higher in dysplasia and CAC, respectively, compared with areas of mild colitis in patient tissue sections. The signal-to-noise ratios for dysplastic lesions seen by endoscopy in Il10−/− mice were 5.2 ± 1.3 (P = 0.0001). Lesions with increased NIRF emissions were classified as raised or flat dysplasia, lymphoid tissue, and ulcers. Using images collected by endoscopy, a receiver operating characteristic curve for correctly diagnosing dysplasia was calculated. The area under the curve was 0.927. At a cutoff of 1000 mean fluorescence intensity, the sensitivity and specificity for detecting dysplasia were 100% and 83%, respectively. Analysis revealed that focally enhanced NIRF emissions derived from increased numbers of infiltrating myeloid-derived suppressor cells and macrophages with equivalent cathepsin activity. Conclusions:These studies indicate that cathepsin substrate-based probe imaging correctly identifies dysplastic foci within chronically inflamed colons. Combined white light and NIRF endoscopy presents unique advantages that may increase sensitivity and specificity of surveillance colonoscopy in patients with CAC.


Alcoholism: Clinical and Experimental Research | 2013

Ethanol‐Induced Mast Cell‐Mediated Inflammation Leads to Increased Susceptibility of Intestinal Tumorigenesis in the APCΔ468 Min Mouse Model of Colon Cancer

Andre L. Wimberly; Christopher B. Forsyth; Mohammad W. Khan; Alan D. Pemberton; Khashayarsha Khazaie; Ali Keshavarzian

BACKGROUND Chronic and frequent alcohol (ethanol [EtOH]) intake has been associated with an increased incidence of several types of cancers including breast, mouth, throat, esophageal, stomach, and colorectal (CRC). The underlying mechanism of this deleterious carcinogenic effect of alcohol has not been clearly established but inflammation may be 1 unifying feature of these cancers. We have recently shown that intestinal mast cells play a central role in intestinal carcinogenesis. In this study, we tested our hypothesis that mast cell-mediated inflammation is 1 underlying mechanism by which chronic alcohol promotes intestinal tumorigenesis. METHODS APC(Δ468) mice were fed either an alcohol-containing Nanji liquid diet or isocaloric dextrose-containing Nanji diet for 10 weeks and then sacrificed to collect small and large intestine samples. Assessments of tumor number and size as well as mast cell number and mast cell activity and histology score for invasion were compared between Control (dextrose-fed) and alcohol-fed APC(∆468) mice. The effect of alcohol on mast cell-mediated tumor migration was also assessed using an in vitro migration assay. RESULTS Alcohol feeding increased both polyp number and size within both the small and the large intestines of APC(∆468) mice. Only alcohol-fed mice showed evidence of tumor invasion. Chronic alcohol feeding also resulted in an increased mast cell number and activity in tumor stroma and invading borders. In vitro migration assay showed that alcohol significantly increases mast cell-mediated tumor migration in vitro. CONCLUSIONS Our data show that chronic alcohol intake promotes: (i) intestinal tumorigenesis and tumor invasion in genetically susceptible mice; (ii) increases in polyp-associated mast cells; and (iii) mast cell-mediated tumor migration in vitro. Both our in vivo and in vitro studies suggest that mast cell-mediated inflammation could be 1 mechanism by which alcohol promotes carcinogenesis.

Collaboration


Dive into the Mohammad W. Khan's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mengxi Tian

San Diego State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Praveen Bere

Northwestern University

View shared research outputs
Researchain Logo
Decentralizing Knowledge