Network


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

Hotspot


Dive into the research topics where Michele B. Weiss is active.

Publication


Featured researches published by Michele B. Weiss.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Knockin of mutant PIK3CA activates multiple oncogenic pathways

John P. Gustin; Bedri Karakas; Michele B. Weiss; Abde M. Abukhdeir; Josh Lauring; Joseph P. Garay; David Cosgrove; Akina Tamaki; Hiroyuki Konishi; Yuko Konishi; Morassa Mohseni; Grace M. Wang; D. Marc Rosen; Samuel R. Denmeade; Michaela J. Higgins; Michele I. Vitolo; Kurtis E. Bachman; Ben Ho Park

The phosphatidylinositol 3-kinase subunit PIK3CA is frequently mutated in human cancers. Here we used gene targeting to “knock in” PIK3CA mutations into human breast epithelial cells to identify new therapeutic targets associated with oncogenic PIK3CA. Mutant PIK3CA knockin cells were capable of epidermal growth factor and mTOR-independent cell proliferation that was associated with AKT, ERK, and GSK3β phosphorylation. Paradoxically, the GSK3β inhibitors lithium chloride and SB216763 selectively decreased the proliferation of human breast and colorectal cancer cell lines with oncogenic PIK3CA mutations and led to a decrease in the GSK3β target gene CYCLIN D1. Oral treatment with lithium preferentially inhibited the growth of nude mouse xenografts of HCT-116 colon cancer cells with mutant PIK3CA compared with isogenic HCT-116 knockout cells containing only wild-type PIK3CA. Our findings suggest GSK3β is an important effector of mutant PIK3CA, and that lithium, an FDA-approved therapy for bipolar disorders, has selective antineoplastic properties against cancers that harbor these mutations.


Cancer Research | 2012

TWIST1 is an ERK1/2 effector that promotes invasion and regulates MMP-1 expression in human melanoma cells

Michele B. Weiss; Ethan V. Abel; Melanie M. Mayberry; Kevin J. Basile; Adam C. Berger; Andrew E. Aplin

Tumor cells often use developmental processes to progress toward advanced disease. The E-box transcription factor TWIST1 is essential to epithelial-mesenchymal transition (EMT) and cell migration in the developing neural crest. In melanoma, which derives from the neural crest cell lineage, enhanced TWIST1 expression has been linked to worse clinical prognosis. However, mechanisms underlying TWIST1 expression and whether aberrant TWIST1 levels promote steps in melanoma progression remain unknown. Here, we report that elevated TWIST1 mRNA/protein expression is dependent on extracellular signal-regulated kinase (ERK)1/2 signaling, which is hyperactive in the majority of melanomas. We show that TWIST1 protein levels are especially high in melanoma cell lines generated from invasive, premetastatic stage tumors. Furthermore, TWIST1 expression is required and sufficient to promote invasion through Matrigel and spheroid outgrowth in three-dimensional dermal-mimetic conditions. Alterations to spheroid outgrowth were not as a result of altered cell death, cell-cycle profile, or paradigm EMT protein changes. Importantly, we identify matrix metalloproteinase-1 (MMP-1) as a novel downstream target of TWIST1. We have determined that TWIST1 acts, in a dose-dependent manner, as a mediator between hyperactive ERK1/2 signaling and regulation of MMP-1 transcription. Together, these studies mechanistically show a previously unrecognized interplay between ERK1/2, TWIST1, and MMP-1 that is likely significant in the progression of melanoma toward metastasis.


Cancer Research | 2009

Deletion of PTEN Promotes Tumorigenic Signaling, Resistance to Anoikis, and Altered Response to Chemotherapeutic Agents in Human Mammary Epithelial Cells

Michele I. Vitolo; Michele B. Weiss; Marta Szmacinski; Khola Tahir; Todd Waldman; Ben Ho Park; Stuart S. Martin; David J. Weber; Kurtis E. Bachman

Many cancers, including breast cancer, harbor loss-of-function mutations in the catalytic domain of phosphatase and tensin homologue deleted on chromosome 10 (PTEN) or have reduced PTEN expression through loss of heterozygosity and/or epigenetic silencing mechanisms. However, specific phenotypic effects of PTEN inactivation in human cancer cells remain poorly defined without a direct causal connection between the loss of PTEN function and the development or progression of cancer. To evaluate the biological and clinical relevance of reduced or deleted PTEN expression, a novel in vitro model system was generated using human somatic cell knockout technologies. Targeted homologous recombination allowed for a single and double allelic deletion, which resulted in reduced and deleted PTEN expression, respectively. We determined that heterozygous loss of PTEN in the nontumorigenic human mammary epithelial cell line MCF-10A was sufficient for activation of the phosphoinositide 3-kinase/AKT and mitogen-activated protein kinase pathways, whereas the homozygous absence of PTEN expression led to a further increased activation of both pathways. The deletion of PTEN was able to confer growth factor-independent proliferation, which was confirmed by the resistance of the PTEN(-/-) MCF-10A cells to small-molecule inhibitors of the epidermal growth factor receptor. However, neither heterozygous nor homozygous loss of PTEN expression was sufficient to promote anchorage-independent growth, but the loss of PTEN did confer apoptotic resistance to cell rounding and matrix detachment. Finally, MCF-10A cells with the reduction or loss of PTEN showed increased susceptibility to the chemotherapeutic drug doxorubicin but not paclitaxel.


Journal of Investigative Dermatology | 2013

The TWEAK Receptor Fn14 Is a Therapeutic Target in Melanoma: Immunotoxins Targeting Fn14 Receptor for Malignant Melanoma Treatment

Hong Zhou; Suhendan Ekmekcioglu; John W. Marks; Khalid A. Mohamedali; Kaushal Asrani; Keeley K. Phillips; Sharron A.N. Brown; Emily Cheng; Michele B. Weiss; Walter N. Hittelman; Nhan L. Tran; Hideo Yagita; Jeffrey A. Winkles; Michael G. Rosenblum

Fn14, the cell surface receptor for TWEAK, is over-expressed in various human solid tumor types and can be a negative prognostic indicator. We detected Fn14 expression in ~60% of the melanoma cell lines we tested, including both B-Raf WT and B-RafV600E lines. Tumor tissue microarray analysis indicated that Fn14 expression was low in normal skin but elevated in 173/190 (92%) of primary melanoma specimens and in 86/150 (58%) of melanoma metastases tested. We generated both a chemical conjugate composed of the rGel toxin and the anti-Fn14 antibody ITEM-4 (designated ITEM4-rGel) and a humanized, dimeric single-chain antibody of ITEM-4 fused to rGel (designated hSGZ). Both ITEM4-rGel and hSGZ were highly cytotoxic to a panel of different melanoma cell lines. Mechanistic studies showed that both immunotoxins induced melanoma cell necrosis. Also, these immunotoxins could up-regulate the cellular expression of Fn14 and trigger cell signaling events similar to the Fn14 ligand TWEAK. Finally, treatment of mice bearing human melanoma MDA-MB-435 xenografts with either ITEM4-rGel or hSGZ showed significant tumor growth inhibition compared to controls. We conclude that Fn14 is a novel therapeutic target in melanoma and the hSGZ construct appears to warrant further development as a novel therapeutic agent against Fn14-positive melanoma.


Oncogene | 2010

Deletion of p53 in human mammary epithelial cells causes chromosomal instability and altered therapeutic response

Michele B. Weiss; Michele I. Vitolo; Morassa Mohseni; D M Rosen; Samuel R. Denmeade; Ben Ho Park; David J. Weber; Kurtis E. Bachman

The TP53 tumor suppressor gene is the most commonly mutated gene in human cancers. To evaluate the biological and clinical relevance of p53 loss, human somatic cell gene targeting was used to delete the TP53 gene in the non-tumorigenic epithelial cell line, MCF-10A. In all four p53−/− clones generated, cells acquired the capability for epidermal growth factor-independent growth and were defective in appropriate downstream signaling and cell cycle checkpoints in response to DNA damage. Interestingly, p53 loss induced chromosomal instability leading to features of transformation and the selection of clones with varying phenotypes. For example, p53-deficient clones were heterogeneous in their capacity for anchorage-independent growth and invasion. In addition, and of clinical importance, the cohort of p53-null clones showed sensitivity to chemotherapeutic interventions that varied depending not only on the type of chemotherapeutic agent, but also on the treatment schedule. In conclusion, deletion of the TP53 gene from MCF-10A cells eliminated p53 functions, as well as produced p53−/− clones with varying phenotypes possibly stemming from the distinct chromosomal changes observed. Such a model system will be useful to further understand the cancer-specific phenotypic changes that accompany p53 loss, as well as help to provide future treatment strategies for human malignancies that harbor aberrant p53.


Molecular Cancer Research | 2014

FOXD3 modulates migration through direct transcriptional repression of TWIST1 in melanoma.

Michele B. Weiss; Ethan V. Abel; Neda Dadpey; Andrew E. Aplin

The neural crest is a multipotent, highly migratory cell population that gives rise to diverse cell types, including melanocytes. Factors regulating the development of the neural crest and emigration of its cells are likely to influence melanoma metastasis. The transcription factor FOXD3 plays an essential role in premigratory neural crest development and has been implicated in melanoma cell dormancy and response to therapeutics. FOXD3 is downregulated during the migration of the melanocyte lineage from the neural crest, and our previous work supports a role for FOXD3 in suppressing melanoma cell migration and invasion. Alternatively, TWIST1 is known to have promigratory and proinvasive roles in a number of cancers, including melanoma. Using ChIP-seq analysis, TWIST1 was identified as a potential transcriptional target of FOXD3. Mechanistically, FOXD3 directly binds to regions of the TWIST1 gene locus, leading to transcriptional repression of TWIST1 in human mutant BRAF melanoma cells. In addition, depletion of endogenous FOXD3 promotes upregulation of TWIST1 transcripts and protein. Finally, FOXD3 expression leads to a significant decrease in cell migration that can be efficiently reversed by the overexpression of TWIST1. These findings uncover the novel interplay between FOXD3 and TWIST1, which is likely to be important in the melanoma metastatic cascade. Implications: FOXD3 and TWIST1 define distinct subgroups of cells within a heterogeneous tumor. Mol Cancer Res; 12(9); 1314–23. ©2014 AACR.


Journal of Investigative Dermatology | 2010

Paying “Particle” Attention to Novel Melanoma Treatment Strategies

Michele B. Weiss; Andrew E. Aplin

Malignant melanoma remains the deadliest form of skin cancer because of its highly aggressive nature and the lack of effective treatments. Recent investigations into alternative treatment strategies have highlighted the exciting potential of nanoparticles to increase melanoma cell delivery and the efficacy of small interfering RNAs (siRNAs) and pharmacological inhibitors. In this issue, Chen et al. report a new liposomal nanoparticle for c-Myc siRNA delivery, noting it to be highly effective in reducing c-Myc expression and inhibiting melanoma tumor growth in mouse models. This preclinical study underscores the importance of investigating nanoparticle treatment options for chemoresistant melanomas.


Cancer Gene Therapy | 2007

Persistent mismatch repair deficiency following targeted correction of hMLH1

Michele B. Weiss; Michele I. Vitolo; Katja Baerenfaller; Giancarlo Marra; Ben Ho Park; Kurtis E. Bachman

The use of gene therapy to correct mutated or lost gene function for the treatment of human cancers has been an active, yet problematic area of biomedical research. Many technical difficulties, including efficient tissue-specific delivery, integration site specificity and general toxicity, are being addressed. Little is known, however, about the genetic and phenotypic stability that accompanies a successful gene-specific targeting event in a cancer cell. This question was addressed following the creation of a colon cancer cell line in which a mutated hMLH1 gene was corrected via targeted homologous recombination. This correction resulted in the expression of wild-type hMLH1 protein, restoration of the hPMS2 protein and mismatch repair (MMR) proficiency. One of two hMLH1-corrected clones, however, was found to retain defects in MMR activity. These cells continued to express the corrected hMLH1 protein, but had lost expression of another MMR protein, hMSH6. DNA sequence analysis of the hMSH6 gene revealed biallelic expansions of a cytosine repeat region in exon 5 that result in frameshifts leading to premature stop codons. These findings suggest that, similar to acquired drug resistance, the presence of genetically heterogeneous cancer cell populations or acquisition of compensatory mutations can result in ‘resistance’ to gene replacement therapy.


Cancer Research | 2015

Abstract A08: CADM1 is a TWIST1 regulated suppressor of melanoma invasion

Edward J. Hartsough; Michele B. Weiss; Sheera Rosenbaum; Andrew E. Aplin

Melanoma is the deadliest form of skin cancer; however, with early detection prior to metastatic dissemination, patients generally have a good prognosis. Recently, our lab has implicated the transcription factor TWIST1 in the progression of melanoma towards metastasis. In melanoma cells, we found that RAS-RAF-MEK-ERK (ERK1/2 pathway) signaling increases TWIST1 expression, which promotes invasive properties in the dermal microenvironment at least in part by enhancing levels of the matrix metalloproteinase, MMP1. Other TWIST1 regulated targets are poorly described. In this study, we compared expression profiling data from cells expressing shRNA against TWIST1 or cells overexpressing TWIST1 in order to determine TWIST1 regulated genes. KEGG and GO analysis revealed that TWIST1 is responsible for regulating a number of genes involved in cellular adhesion. We found that TWIST1 levels inversely correlate with levels of cell adhesion molecule 1 (CADM1) (NECL-2, IGSF4, TSLC1, SynCAM). Chromatin immunoprecipitation (ChIP) studies and promoter assays demonstrate that TWIST1 physically interacts with CADM1 promoter and this is associated with reduced CADM1 levels. Additionally, CADM1 expression is inversely associated ERK1/2 signaling and TWIST1 expression. Modulation of cellular CADM1 levels does not seem to affect proliferations rates however, overexpression of CADM1 augments cell-cell interaction, and cell aggregation. Furthermore, exogenous CADM1 inhibited serum directed migration and invasion through matrigel coated boyden chambers, while knockdown of CADM1 was associated with an enhancement in the migratory and invasion properties. Taken together, these data provide evidence that CADM1 is negatively regulated by TWIST1, and may act as a suppressor of melanoma invasion. Citation Format: Edward J. Hartsough, Michele B. Weiss, Curtis H. Kugel, Sheera R. Rosenbaum, Andrew E. Aplin. CADM1 is a TWIST1 regulated suppressor of melanoma invasion. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 4089. doi:10.1158/1538-7445.AM2015-4089


Cancer Research | 2012

Abstract 3866: TWEAK receptor (Fn14) Is a novel target in melanoma: Characterization of unique targeted therapeutics

Hong Zhou; Suhendan Ekmekcioglu; John W. Marks; Kaushal Asrani; Keeley K. Phillips; Sharron A.N. Brown; Emily Cheng; Michele B. Weiss; Walter N. Hittelman; Nhan L. Tran; Hideo Yagita; Jeffrey A. Winkles; Michael G. Rosenblum

Proceedings: AACR 103rd Annual Meeting 2012‐‐ Mar 31‐Apr 4, 2012; Chicago, IL Previous studies have indicated that Fn14, the cell surface receptor for the cytokine TWEAK, is over-expressed in multiple human solid tumor types, including brain, breast, and lung cancers, and overexpression can be a negative prognostic indicator. We analyzed a series of melanoma cell lines and tumor tissue microarrays and detected Fn14 expression in ∼60% of the melanoma cell lines, including both B-Raf WT and B-Raf V600E lines. Fn14 expression was elevated in 178/190 (93.6%) of primary melanoma specimens and in 87/150 (58%) of melanoma metastases tested. Fn14 expression was not elevated in normal skin tissues. Initial development and characterization of an immunoconjugate designated ITEM4-rGel targeting Fn14 receptor has been published (Zhou et al., Mol. Cancer Ther. 10:1276 (2011)). We have now developed an Fn14-targeted immunotoxin more suitable for long-term clinical use. Specifically, we generated a humanized, dimeric single-chain version of ITEM-4 and fused this scFv to rGel. The resulting anti-Fn14 immunotoxin, designated hSGZ, bound to Fn14 with a Kd of ∼1.4 nM as determined by Biacore analysis. Confocal immunofluoresence studies showed that hSGZ specifically and rapidly (within 2 hrs) internalized into Fn14-expressing MDA-MB-435 melanoma cells. Cytotoxicity studies showed that hSGZ was highly cytotoxic to a panel of different melanoma cell lines (IC50 ranged from 0.1 pM to 1.1 nM) and was 2.2 to 2.8 ×105 fold more potent than free rGel. Treatment of cells expressing the multidrug resistance protein MDR1 showed no cross-resistance to the fusion construct in vitro. When hSGZ was combined with 5-FU, cisplatin, doxorubicin, etoposide or dacarbazine, we found an additive effect on melanoma cell growth inhibition. Mechanistic studies showed that hSGZ induced melanoma cell death consistent with a necrotic mechanism. Additionally, Fn14-targeted immunotoxins increased Fn14 expression and triggered cell signaling events similar to those induced by the TWEAK ligand. Finally, treatment of mice bearing human melanoma MDA-MB-435 and breast MDA-MB-231 xenografts with either ITEM4-rGel or hSGZ showed significant tumor growth inhibition compared to controls (P< 0.05). Fn14 appears to be an excellent new target for melanoma and the Fn14-targeting construct hSGZ appears to warrant further development as a novel therapeutic agent against Fn14-positive tumors. Melanoma lines appear to be the most sensitive tumor type tested but the reasons for this are unclear. Additional studies are in progress to investigate the biodistribution and pharmacokinetics of hSGZ in tumor-bearing mice. This work was conducted, in part, by the Clayton Foundation for Research (MGR); and supported by NIH grant NS055126 (JAW) and DOD Breast Cancer Concept Award BC086135 (JAW). Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 3866. doi:1538-7445.AM2012-3866

Collaboration


Dive into the Michele B. Weiss's collaboration.

Top Co-Authors

Avatar

Andrew E. Aplin

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ben Ho Park

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ethan V. Abel

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Emily Cheng

University of Maryland

View shared research outputs
Top Co-Authors

Avatar

Hong Zhou

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge