3,516 results on '"Tumor initiation"'
Search Results
2. A dataset of chromosomal instability gene signature scores in normal and cancer cells from the human breast.
- Author
-
Baba, Shahnawaz, Labhsetwar, Shreyas, Klemke, Richard, and Desgrosellier, Jay
- Subjects
Breast cancer subtype ,Cancer stem cell ,Chromosomal missegregation ,Signaling state ,Stress tolerance ,Tumor initiation - Abstract
These data show the relative amount of chromosomal instability (CIN) in a diverse array of human breast cell types, including non-transformed mammary epithelial cells as well as cancer cell lines. Additional data is also provided from human embryonic and mesenchymal stem cells. To produce this dataset, we compared a published chromosomal instability gene signature against publicly available datasets containing gene expression information for each cell. We then analyzed these data with the Python GSEAPY software package to provide a CIN enrichment score. These data are useful for comparing the relative amounts of CIN in different breast cell types. This includes cells representing the major clinical (ER/PR+, HER2+ & Triple-negative) as well as intrinsic breast cancer subtypes (Luminal B, HER2+, Basal-like and Claudin-low). Our dataset has a great potential for re-use given the recent surge in interest surrounding the role of CIN in breast cancer. The large size of the dataset, coupled with the diversity of the cell types represented, provides numerous possibilities for future comparisons.
- Published
- 2023
3. The dual role of SOX17 in the occurrence and development of early colorectal cancer
- Author
-
Xinming Su, Jianqiao Shentu, Ruixiu Chen, and Shiwei Duan
- Subjects
SOX17 ,Colorectal cancer ,Immune evasion ,Tumor initiation ,Therapeutic targets ,Medicine - Published
- 2024
- Full Text
- View/download PDF
4. Breast cancer stem cells tolerate chromosomal instability during tumor progression via c-Jun/AXL stress signaling
- Author
-
Baba, Shahnawaz A, Sun, Qi, Mugisha, Samson, Labhsetwar, Shreyas, Klemke, Richard, and Desgrosellier, Jay S
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Prevention ,Cancer ,Breast Cancer ,Stem Cell Research ,Aetiology ,2.1 Biological and endogenous factors ,Breast cancer ,Stem cell activation ,Chromosomal missegregation ,Stress tolerance ,Tumor initiation - Abstract
Chromosomal instability (CIN) is critical for tumor evolution, yet its relationship with stemness is unclear. Here, we describe CIN as a key stress induced during tumor initiation that is uniquely tolerated by breast cancer stem cells in an activated signaling state (aCSCs). While we noted elevated CIN specifically in tumors from aCSCs, this was not intrinsic to these cells, as baseline levels were similar to non-stem cell types. This suggests that CIN is induced during tumor initiation, and that aCSCs can better tolerate this stress. Further, this increased CIN may be transient, as it was only in low-burden aCSC tumors, with levels diminishing in more established disease. Phospho-array profiling revealed specific activation of c-Jun stress signaling in aCSCs, which we hypothesized could induce genes responsible for CIN tolerance. Indeed, we identified AXL as a c-Jun dependent gene enriched in aCSCs that enhances resistance to this stress. Thus, CIN tolerance mediated by c-Jun/AXL signaling may be a defining feature of stemness, contributing to breast cancer progression.
- Published
- 2023
5. Novel Insights into the Initiation, Evolution, and Progression of Multiple Myeloma by Multi-Omics Investigation.
- Author
-
Gong, Lixin, Qiu, Lugui, and Hao, Mu
- Subjects
- *
DISEASE progression , *BIOLOGICAL evolution , *GENETIC mutation , *POLYPHARMACY , *DRUG resistance , *NEOPLASTIC cell transformation , *DISEASE relapse , *HEMATOLOGIC malignancies , *MULTIPLE myeloma , *CELL lines - Abstract
Simple Summary: The integration of genomics, transcriptomics, epigenomics and proteomics broad our understanding of the disease. In this review, we generally introduce the progress in single-cell multi-omics technologies. We summarize the novel insights into the initiation, evolution, and progression of multiple myeloma when using diverse multi-omics sequencing tools. We intend to discuss the challenges and future perspectives of omics in myeloma. Based on these recent omics findings in myeloma, the advanced single-cell multi-omics technologies allow the researchers to acquire both the integral overview of the tumor environment and also the sophisticated internal contexture of the tumor landscape. The evolutionary history of multiple myeloma (MM) includes malignant transformation, followed by progression to pre-malignant stages and overt malignancy, ultimately leading to more aggressive and resistant forms. Over the past decade, large effort has been made to identify the potential therapeutic targets in MM. However, MM remains largely incurable. Most patients experience multiple relapses and inevitably become refractory to treatment. Tumor-initiating cell populations are the postulated population, leading to the recurrent relapses in many hematological malignancies. Clonal evolution of tumor cells in MM has been identified along with the disease progression. As a consequence of different responses to the treatment of heterogeneous MM cell clones, the more aggressive populations survive and evolve. In addition, the tumor microenvironment is a complex ecosystem which plays multifaceted roles in supporting tumor cell evolution. Emerging multi-omics research at single-cell resolution permits an integrative and comprehensive profiling of the tumor cells and microenvironment, deepening the understanding of biological features of MM. In this review, we intend to discuss the novel insights into tumor cell initiation, clonal evolution, drug resistance, and tumor microenvironment in MM, as revealed by emerging multi-omics investigations. These data suggest a promising strategy to unravel the pivotal mechanisms of MM progression and enable the improvement in treatment, both holistically and precisely. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. Breast cancer stem cells generate immune-suppressive T regulatory cells by secreting TGFβ to evade immune-elimination.
- Author
-
Mukherjee, Sumon, Chakraborty, Sourio, Basak, Udit, Pati, Subhadip, Dutta, Apratim, Dutta, Saikat, Roy, Dia, Banerjee, Shruti, Ray, Arpan, Sa, Gaurisankar, and Das, Tanya
- Subjects
REGULATORY T cells ,CANCER stem cells ,T cells ,BREAST cancer ,CANCER cells - Abstract
Cancer stem cells (CSCs), being the primary contributors in tumor initiation, metastasis, and relapse, ought to have seminal roles in evasion of immune surveillance. Tumor-promoting CD4
+ CD25+ FOXP3+ T-regulatory cells (Tregs) have been described to abolish host defense mechanisms by impeding the activities of other immune cells including effector T cells. However, whether CSCs can convert effector T cells to immune-suppressive Treg subset, and if yes, the mechanism underlying CSC-induced Treg generation, are limitedly studied. In this regard, we observed a positive correlation between breast CSC and Treg signature markers in both in-silico and immunohistochemical analyses. Mirroring the conditions during tumor initiation, low number of CSCs could successfully generate CD4+ CD25+ FOXP3+ Treg cells from infiltrating CD4+ T lymphocytes in a contact-independent manner. Suppressing the proliferation potential as well as IFNγ production capacity of effector T cells, these Treg cells might be inhibiting antitumor immunity, thereby hindering immune-elimination of CSCs during tumor initiation. Furthermore, unlike non-stem cancer cells (NSCCs), CSCs escaped doxorubicin-induced apoptosis, thus constituting major surviving population after three rounds of chemotherapy. These drug-survived CSCs were also able to generate CD4+ CD25+ FOXP3+ Treg cells. Our search for the underlying mechanism further unveiled the role of CSC-shed immune-suppressive cytokine TGFβ, which was further increased by chemotherapy, in generating tumor Treg cells. In conclusion, during initiation as well as after chemotherapy, when NSCCs are not present in the tumor microenvironment, CSCs, albeit present in low numbers, generate immunosuppressive CD4+ CD25+ FOXP3+ Treg cells in a contact-independent manner by shedding high levels of immune-suppressive Treg-polarizing cytokine TGFβ, thus escaping immune-elimination and initiating the tumor or causing tumor relapse. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
7. Immune evasion by cancer stem cells ensures tumor initiation and failure of immunotherapy
- Author
-
Sourio Chakraborty, Sumon Mukherjee, Udit Basak, Subhadip Pati, Apratim Dutta, Saikat Dutta, Subhanki Dhar, Tania Sarkar, Aharna Guin, Gaurisankar Sa, and Tanya Das
- Subjects
antitumor immunity ,cancer stem cells ,cancer stem cell sensitizing therapy ,metastasis ,suppressive immune cells ,tumor initiation ,tumor-microenvironment ,tumor relapse ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Cancer stem cells (CSCs) are a small subpopulation of cells that drive the formation and progression of tumors. However, during tumor initiation, how CSCs communicate with neighbouring immune cells to overcome the powerful immune surveillance barrier in order to form, spread, and maintain the tumor, remains poorly understood. It is, therefore, absolutely necessary to understand how a small number of tumor-initiating cells (TICs) survive immune attack during (a) the “elimination phase” of “tumor immune-editing”, (b) the establishment of regional or distant tumor after metastasis, and (c) recurrence after therapy. Mounting evidence suggests that CSCs suppress the immune system through a variety of distinct mechanisms that ensure the survival of not only CSCs but also non-stem cancer cells (NSCCs), which eventually form the tumor mass. In this review article, the mechanisms via which CSCs change the immune landscape of the tissue of origin, which contains macrophages, dendritic cells (DCs), myeloid-derived suppressor cells (MDSCs), natural killer (NK) cells, and tumor-infiltrating lymphocytes, in favour of tumorigenesis were discussed. The failure of cancer immunotherapy might also be explained by such interaction between CSCs and immune cells. This review will shed light on the critical role of CSCs in tumor immune evasion and emphasize the importance of CSC-targeted immunotherapy as a cutting-edge technique for battling cancer by restricting communication between immune cells and CSCs.
- Published
- 2023
- Full Text
- View/download PDF
8. The microRNA target site profile is a novel biomarker in the immunotherapy response
- Author
-
Yulong Bai, Yujia Li, Yidi Qin, Xinshuo Yang, George C. Tseng, Soyeon Kim, and Hyun Jung Park
- Subjects
RNA-level regulation ,microRNA binding mechanism ,tumor initiation ,cancer biology ,biomarker ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
MicroRNAs (miRNAs) bind on the 3′ untranslated region (3′UTR) of messenger RNAs (mRNAs) and regulate mRNA expression in physiological and pathological conditions, including cancer. Thus, studies have identified miRNAs as potential biomarkers by correlating the miRNA expression with the expression of important mRNAs and/or clinical outcomes in cancers. However, tumors undergo pervasive 3′UTR shortening/lengthening events through alternative polyadenylation (APA), which varies the number of miRNA target sites in mRNA, raising the number of miRNA target sites (numTS) as another important regulatory axis of the miRNA binding effects. In this study, we developed the first statistical method, BIOMATA-APA, to identify predictive miRNAs based on numTS features. Running BIOMATA-APA on The Cancer Genome Atlas (TCGA) and independent cohort data both with immunotherapy and no immunotherapy, we demonstrated for the first time that the numTS feature 1) distinguishes different cancer types, 2) predicts tumor proliferation and immune infiltration status, 3) explains more variation in the proportion of tumor-infiltrating immune cells, 4) predicts response to immune checkpoint blockade (ICB) therapy, and 5) adds prognostic power beyond clinical and miRNA expression. To the best of our knowledge, this is the first pan-cancer study to systematically demonstrate numTS as a novel type of biomarker representing the miRNA binding effects underlying tumorigenesis and pave the way to incorporate miRNA target sites for miRNA biomarker identification. Another advantage of examining the miRNA binding effect using numTS is that it requires only RNA-Seq data, not miRNAs, thus resulting in high power in the miRNA biomarker identification.
- Published
- 2023
- Full Text
- View/download PDF
9. A dataset of chromosomal instability gene signature scores in normal and cancer cells from the human breast
- Author
-
Shahnawaz A. Baba, Shreyas Labhsetwar, Richard Klemke, and Jay S. Desgrosellier
- Subjects
Cancer stem cell ,Chromosomal missegregation ,Signaling state ,Stress tolerance ,Breast cancer subtype ,Tumor initiation ,Computer applications to medicine. Medical informatics ,R858-859.7 ,Science (General) ,Q1-390 - Abstract
These data show the relative amount of chromosomal instability (CIN) in a diverse array of human breast cell types, including non-transformed mammary epithelial cells as well as cancer cell lines. Additional data is also provided from human embryonic and mesenchymal stem cells. To produce this dataset, we compared a published chromosomal instability gene signature against publicly available datasets containing gene expression information for each cell. We then analyzed these data with the Python GSEAPY software package to provide a CIN enrichment score. These data are useful for comparing the relative amounts of CIN in different breast cell types. This includes cells representing the major clinical (ER/PR+, HER2+ & Triple-negative) as well as intrinsic breast cancer subtypes (Luminal B, HER2+, Basal-like and Claudin-low). Our dataset has a great potential for re-use given the recent surge in interest surrounding the role of CIN in breast cancer. The large size of the dataset, coupled with the diversity of the cell types represented, provides numerous possibilities for future comparisons.
- Published
- 2023
- Full Text
- View/download PDF
10. Upregulation of fibronectin and its integrin receptors - an adaptation to isolation stress that facilitates tumor initiation.
- Author
-
Chengsheng Wu, Weis, Sara M., and Cheresh, David A.
- Subjects
- *
CELL receptors , *G protein coupled receptors , *INTEGRINS , *FIBRONECTINS , *CELL separation , *TUMOR proteins , *LYSOPHOSPHOLIPIDS - Abstract
Tumor initiation at either primary or metastatic sites is an inefficient process in which tumor cells must fulfill a series of conditions. One critical condition involves the ability of individual tumor-initiating cells to overcome 'isolation stress', enabling them to survive within harsh isolating microenvironments that can feature nutrient stress, hypoxia, oxidative stress and the absence of a proper extracellular matrix (ECM). In response to isolation stress, tumor cells can exploit various adaptive strategies to develop stress tolerance and gain stemness features. In this Opinion, we discuss how strategies such as the induction of certain cell surface receptors and deposition of ECM proteins enable tumor cells to endure isolation stress, thereby gaining tumor-initiating potential. As examples, we highlight recent findings from our group demonstrating how exposure of tumor cells to isolation stress upregulates the G-protein-coupled receptor lysophosphatidic acid receptor 4 (LPAR4), its downstream target fibronectin and two fibronectin-binding integrins, α5β1 and αvβ3. These responses create a fibronectin-rich niche for tumor cells, ultimately driving stress tolerance, cancer stemness and tumor initiation. We suggest that approaches to prevent cancer cells from adapting to stress by suppressing LPAR4 induction, blocking its downstream signaling or disrupting fibronectin-integrin interactions hold promise as potential strategies for cancer treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
11. Loss of MXRA8 Delays Mammary Tumor Development and Impairs Metastasis.
- Author
-
Simpson, Kaitlyn E., Staikos, Christina A., Watson, Katrina L., and Moorehead, Roger A.
- Subjects
- *
TRIPLE-negative breast cancer , *CELL migration , *CELL communication , *CHIKUNGUNYA virus , *EXTRACELLULAR matrix , *BREAST - Abstract
Matrix-remodeling-associated protein 8 or MXRA8 is a transmembrane protein that can bind arthritogenic alpha viruses like the Chikungunya virus and provide viral entry into cells. MXRA8 can also interact with integrin β3 and thus possibly regulate cell–cell interactions and binding to the extracellular matrix. While MXRA8 has been associated with reduced survival in patients with colorectal and renal clear cell cancers, the role of MXRA8 in breast cancer remains largely unexplored. Therefore, the aim of this research was to determine the role of MXRA8 in breast cancer by knocking out MXRA8 in the human triple-negative breast cancer cell line MDA-MB-231. The loss of MXRA8 reduced cell proliferation in vitro but had no effect on apoptosis or migration in cultured cells. However, the loss of MXRA8 significantly delayed tumor development and reduced metastatic dissemination to the lungs in a xenograft model. RNA sequencing identified three genes, ADMATS1, TIE1, and BMP2, whose expression were significantly reduced in MXRA8-knockout tumors compared to control tumors. MXRA8 staining of a human breast cancer tissue array revealed higher levels of MXRA8 in primary tumors and metastases of aggressive tumor subtypes (TNBC and HER2+) compared to less aggressive, ER+ breast cancers. Our findings demonstrate for the first time that MXRA8 regulates the progression of human TNBC possibly through influencing the interaction of tumor cells with their microenvironment. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
12. Ezh2 promotes mammary tumor initiation through epigenetic regulation of the Wnt and mTORC1 signaling pathways.
- Author
-
Linshan Liu, Bin Xiao, Hirukaw, Alison, Smith, Harvey W., Dongmei Zuo, Sanguin-Gendreau, Virginie, McCaffrey, Luke, Nam, Alice Jisoo, and Muller, William J.
- Subjects
- *
WNT signal transduction , *CELLULAR signal transduction , *GENETIC regulation , *EPIGENETICS , *POST-translational modification - Abstract
The regulation of gene expression through histone posttranslational modifications plays a crucial role in breast cancer progression. However, the molecular mechanisms underlying the contribution of histone modification to tumor initiation remain unclear. To gain a deeper understanding of the role of the histone modifier Enhancer of Zeste homology 2 (Ezh2) in the early stages of mammary tumor progression, we employed an inducible mammary organoid system bearing conditional Ezh2 alleles that faithfully recapitulates key events of luminal B breast cancer initiation. We showed that the loss of Ezh2 severely impairs oncogene-induced organoid growth, with Ezh2-deficient organoids maintaining a polarized epithelial phenotype. Transcriptomic profiling showed that Ezh2-deficient mammary epithelial cells up-regulated the expression of negative regulators of Wnt signaling and down-regulated genes involved in mTORC1 (mechanistic target of rapamycin complex 1) signaling. We identified Sfrp1, a Wnt signaling suppressor, as an Ezh2 target gene that is derepressed and expressed in Ezh2-deficient epithelium. Furthermore, an analysis of breast cancer data revealed that Sfrp1 expression was associated with favorable clinical outcomes in luminal B breast cancer patients. Finally, we confirmed that targeting Ezh2 impairs mTORC1 activity through an indirect mechanism that up-regulates the expression of the tumor suppressor Pten. These findings indicate that Ezh2 integrates the mTORC1 and Wnt signaling pathways during early mammary tumor progression, arguing that inhibiting Ezh2 or therapeutically targeting Ezh2-dependent programs could be beneficial for the treatment of early-stage luminal B breast cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
13. Breast cancer stem cells tolerate chromosomal instability during tumor progression via c-Jun/AXL stress signaling
- Author
-
Shahnawaz A. Baba, Qi Sun, Samson Mugisha, Shreyas Labhsetwar, Richard Klemke, and Jay S. Desgrosellier
- Subjects
Breast cancer ,Stem cell activation ,Chromosomal missegregation ,Stress tolerance ,Tumor initiation ,Science (General) ,Q1-390 ,Social sciences (General) ,H1-99 - Abstract
Chromosomal instability (CIN) is critical for tumor evolution, yet its relationship with stemness is unclear. Here, we describe CIN as a key stress induced during tumor initiation that is uniquely tolerated by breast cancer stem cells in an activated signaling state (aCSCs). While we noted elevated CIN specifically in tumors from aCSCs, this was not intrinsic to these cells, as baseline levels were similar to non-stem cell types. This suggests that CIN is induced during tumor initiation, and that aCSCs can better tolerate this stress. Further, this increased CIN may be transient, as it was only in low-burden aCSC tumors, with levels diminishing in more established disease. Phospho-array profiling revealed specific activation of c-Jun stress signaling in aCSCs, which we hypothesized could induce genes responsible for CIN tolerance. Indeed, we identified AXL as a c-Jun dependent gene enriched in aCSCs that enhances resistance to this stress. Thus, CIN tolerance mediated by c-Jun/AXL signaling may be a defining feature of stemness, contributing to breast cancer progression.
- Published
- 2023
- Full Text
- View/download PDF
14. GSK-3α/β and MEK inhibitors assist the microenvironment of tumor initiation.
- Author
-
Hassan, Ghmkin, Afify, Said M., Zahra, Maram H., Nawara, Hend M., Kumon, Kazuki, Iwasaki, Yoshiaki, Salomon, David S., Seno, Akimasa, and Seno, Masaharu
- Abstract
Induced pluripotent stem cells (iPSCs) are useful tools for modeling diseases and developing personalized medicine. We have been developing cancer stem cells (CSCs) from iPSCs with conditioned medium (CM) of cancer-derived cells as the mimicry of the microenvironment of tumor initiation. However, the conversion of human iPSCs has not always been efficient with only CM. In this study, human iPSCs reprogrammed from monocytes of healthy volunteers were cultured in a media containing 50% of the CM from human pancreatic cancer derived BxPC3 cells supplemented with a MEK inhibitor (AZD6244) and a GSK-3α/β inhibitor (CHIR99021). The survived cells were assessed for the characteristics of CSCs in vitro and in vivo. As a result, they exhibited CSC phenotypes of self-renewal, differentiation, and malignant tumorigenicity. Primary culture of the malignant tumors of the converted cells exhibited the elevated expression of CSC related genes CD44, CD24 and EPCAM maintaining the expression of stemness genes. In conclusion, the inhibition of GSK-3α/β and MEK and the microenvironment of tumor initiation mimicked by the CM can convert human normal stem cells into CSCs. This study could provide insights into establishing potentially novel personalized cancer models which could help investigate the tumor initiation and screening of personalized therapies on CSCs. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
15. On the Role of Glycolysis in Early Tumorigenesis—Permissive and Executioner Effects.
- Author
-
Marcucci, Fabrizio and Rumio, Cristiano
- Subjects
- *
GLYCOLYSIS , *NEOPLASTIC cell transformation , *STRAINS & stresses (Mechanics) , *EPITHELIAL-mesenchymal transition , *DNA repair , *EXECUTIONS & executioners - Abstract
Reprogramming energy production from mitochondrial respiration to glycolysis is now considered a hallmark of cancer. When tumors grow beyond a certain size they give rise to changes in their microenvironment (e.g., hypoxia, mechanical stress) that are conducive to the upregulation of glycolysis. Over the years, however, it has become clear that glycolysis can also associate with the earliest steps of tumorigenesis. Thus, many of the oncoproteins most commonly involved in tumor initiation and progression upregulate glycolysis. Moreover, in recent years, considerable evidence has been reported suggesting that upregulated glycolysis itself, through its enzymes and/or metabolites, may play a causative role in tumorigenesis, either by acting itself as an oncogenic stimulus or by facilitating the appearance of oncogenic mutations. In fact, several changes induced by upregulated glycolysis have been shown to be involved in tumor initiation and early tumorigenesis: glycolysis-induced chromatin remodeling, inhibition of premature senescence and induction of proliferation, effects on DNA repair, O-linked N-acetylglucosamine modification of target proteins, antiapoptotic effects, induction of epithelial–mesenchymal transition or autophagy, and induction of angiogenesis. In this article we summarize the evidence that upregulated glycolysis is involved in tumor initiation and, in the following, we propose a mechanistic model aimed at explaining how upregulated glycolysis may play such a role. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
16. Defining function of wild-type and three patient-specific TP53 mutations in a zebrafish model of embryonal rhabdomyosarcoma
- Author
-
Jiangfei Chen, Kunal Baxi, Amanda E Lipsitt, Nicole Rae Hensch, Long Wang, Prethish Sreenivas, Paulomi Modi, Xiang Ru Zhao, Antoine Baudin, Daniel G Robledo, Abhik Bandyopadhyay, Aaron Sugalski, Anil K Challa, Dias Kurmashev, Andrea R Gilbert, Gail E Tomlinson, Peter Houghton, Yidong Chen, Madeline N Hayes, Eleanor Y Chen, David S Libich, and Myron S Ignatius
- Subjects
zebrafish ,TP53 ,rhabdomyosarcoma ,tumor initiation ,mutant tp53 ,rare varients ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
In embryonal rhabdomyosarcoma (ERMS) and generally in sarcomas, the role of wild-type and loss- or gain-of-function TP53 mutations remains largely undefined. Eliminating mutant or restoring wild-type p53 is challenging; nevertheless, understanding p53 variant effects on tumorigenesis remains central to realizing better treatment outcomes. In ERMS, >70% of patients retain wild-type TP53, yet mutations when present are associated with worse prognosis. Employing a kRASG12D-driven ERMS tumor model and tp53 null (tp53-/-) zebrafish, we define wild-type and patient-specific TP53 mutant effects on tumorigenesis. We demonstrate that tp53 is a major suppressor of tumorigenesis, where tp53 loss expands tumor initiation from 97% of animals. Characterizing three patient-specific alleles reveals that TP53C176F partially retains wild-type p53 apoptotic activity that can be exploited, whereas TP53P153Δ and TP53Y220C encode two structurally related proteins with gain-of-function effects that predispose to head musculature ERMS. TP53P153Δ unexpectedly also predisposes to hedgehog-expressing medulloblastomas in the kRASG12D-driven ERMS-model.
- Published
- 2023
- Full Text
- View/download PDF
17. Lessons learned from SMAD4 loss in squamous cell carcinomas
- Author
-
Hernandez, Ariel L, Young, Christian D, Wang, Jing H, and Wang, Xiao‐Jing
- Subjects
Cancer ,Genetics ,Human Genome ,Aetiology ,2.1 Biological and endogenous factors ,Animals ,Biomarkers ,Tumor ,Carcinoma ,Squamous Cell ,DNA Repair ,Epithelial Cells ,Genes ,Tumor Suppressor ,Humans ,Signal Transduction ,Smad4 Protein ,Tumor Microenvironment ,prevalence ,SCCs ,SMAD4 loss ,tumor initiation ,tumor progression ,Oncology and Carcinogenesis ,Oncology & Carcinogenesis - Abstract
SMAD4 is a potent tumor suppressor and a central mediator of the TGFß signaling pathway. SMAD4 genetic loss is frequent in squamous cell carcinomas (SCCs). Reports of SMAD4 expression in SCCs vary significantly possibly due to inter-tumor heterogeneity or technical reasons. SMAD4 loss is an initiation event for SCCs. In tumor epithelial cells, SMAD4 loss causes increased proliferation, decreased apoptosis, and "Brca-like" genomic instability associated with DNA repair defects. SMAD4 loss also plays a role in the expansion of cancer stem cells. Epithelial SMAD4 loss causes overexpression of TGFß that is released into the tumor microenvironment and contributes to SCC progression through proinflammatory and immune evasive mechanisms. SMAD4 loss, while not a direct therapeutic target, is associated with multiple targetable pathways that require further therapeutic studies. Altogether, SMAD4 loss is a potential biomarker in SCCs that should be further studied for its values in prognostic and therapeutic predictions. Such information will potentially guide future biomarker-driven clinical trial designs and improve SCC patient outcomes.
- Published
- 2019
18. Cellular Plasticity and Heterotypic Interactions during Breast Morphogenesis and Cancer Initiation.
- Author
-
Ingthorsson, Saevar, Traustadottir, Gunnhildur Asta, and Gudjonsson, Thorarinn
- Subjects
- *
MORPHOGENESIS , *DISEASE progression , *CELL differentiation , *IMMUNOGLOBULINS , *CELL physiology , *MENSTRUATION , *APOPTOSIS , *STROMAL cells , *CELL proliferation , *EPITHELIAL cells , *HEMATOPOIETIC stem cells , *BREAST tumors - Abstract
Simple Summary: This review aims to discuss the structure, function and dynamics of the breast gland and how changes to the function of the breast's cells can lead to different types of cancer. The human breast gland is a unique organ as most of its development occurs postnatally between menarche and menopause, a period ranging from 30 to 40 years. During this period, the monthly menstruation cycle drives the mammary gland through phases of cell proliferation, differentiation, and apoptosis, facilitated via a closely choreographed interaction between the epithelial cells and the surrounding stroma preparing the gland for pregnancy. If pregnancy occurs, maximal differentiation is reached to prepare for lactation. After lactation, the mammary gland involutes to a pre-pregnant state. These cycles of proliferation, differentiation, and involution necessitate the presence of epithelial stem cells that give rise to progenitor cells which differentiate further into the luminal and myoepithelial lineages that constitute the epithelial compartment and are responsible for the branching structure of the gland. Maintaining homeostasis and the stem cell niche depends strongly on signaling between the stem and progenitor cells and the surrounding stroma. Breast cancer is a slowly progressing disease whose initiation can take decades to progress into an invasive form. Accumulating evidence indicates that stem cells and/or progenitor cells at different stages, rather than terminally differentiated cells are the main cells of origin for most breast cancer subgroups. Stem cells and cancer cells share several similarities such as increased survival and cellular plasticity which is reflected in their ability to switch fate by receiving intrinsic and extrinsic signals. In this review, we discuss the concept of cellular plasticity in normal breast morphogenesis and cancer, and how the stromal environment plays a vital role in cancer initiation and progression. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
19. Epithelial aPKC deficiency leads to stem cell loss preceding metaplasia in colorectal cancer initiation.
- Author
-
Kinoshita, Hiroto, Martinez-Ordoñez, Anxo, Cid-Diaz, Tania, Han, Qixiu, Duran, Angeles, Muta, Yu, Zhang, Xiao, Linares, Juan F., Nakanishi, Yuki, Kasashima, Hiroaki, Yashiro, Masakazu, Maeda, Kiyoshi, Albaladejo-Gonzalez, Ana, Torres-Moreno, Daniel, García-Solano, José, Conesa-Zamora, Pablo, Inghirami, Giorgio, Diaz-Meco, Maria T., and Moscat, Jorge
- Subjects
- *
CANCER stem cells , *INTESTINAL mucosa , *STEM cells , *CELL populations , *PRECANCEROUS conditions - Abstract
The early mechanisms of spontaneous tumor initiation that precede malignancy are largely unknown. We show that reduced aPKC levels correlate with stem cell loss and the induction of revival and metaplastic programs in serrated- and conventional-initiated premalignant lesions, which is perpetuated in colorectal cancers (CRCs). Acute inactivation of PKCλ/ι in vivo and in mouse organoids is sufficient to stimulate JNK in non-transformed intestinal epithelial cells (IECs), which promotes cell death and the rapid loss of the intestinal stem cells (ISCs), including those that are LGR5+. This is followed by the accumulation of revival stem cells (RSCs) at the bottom of the crypt and fetal-metaplastic cells (FMCs) at the top, creating two spatiotemporally distinct cell populations that depend on JNK-induced AP-1 and YAP. These cell lineage changes are maintained during cancer initiation and progression and determine the aggressive phenotype of human CRC, irrespective of their serrated or conventional origin. [Display omitted] • aPKC deficiency acutely drives intrinsic stem cell loss in the intestinal epithelium • Stem cells are lost before metaplasia and in aPKC-low dysplasia and CRC • Revival stem and fetal metaplasia cells are two spatiotemporally distinct populations • JNK activation by low aPKC leads to stem cell loss and metaplasia through AP-1/YAP Kinoshita and Martinez-Ordoñez et al. demonstrate that aPKC loss is an early event in the initiation of colorectal cancer through the conventional and serrated pathways. This is associated with impaired intestinal stem cells, preceding the activation of the revival and metaplastic cell programs through a JNK-driven AP-1/YAP cascade. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
20. Loss of MXRA8 Delays Mammary Tumor Development and Impairs Metastasis
- Author
-
Kaitlyn E. Simpson, Christina A. Staikos, Katrina L. Watson, and Roger A. Moorehead
- Subjects
TNBC ,MXRA8 ,metastasis ,tumor initiation ,xenograft model ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
Matrix-remodeling-associated protein 8 or MXRA8 is a transmembrane protein that can bind arthritogenic alpha viruses like the Chikungunya virus and provide viral entry into cells. MXRA8 can also interact with integrin β3 and thus possibly regulate cell–cell interactions and binding to the extracellular matrix. While MXRA8 has been associated with reduced survival in patients with colorectal and renal clear cell cancers, the role of MXRA8 in breast cancer remains largely unexplored. Therefore, the aim of this research was to determine the role of MXRA8 in breast cancer by knocking out MXRA8 in the human triple-negative breast cancer cell line MDA-MB-231. The loss of MXRA8 reduced cell proliferation in vitro but had no effect on apoptosis or migration in cultured cells. However, the loss of MXRA8 significantly delayed tumor development and reduced metastatic dissemination to the lungs in a xenograft model. RNA sequencing identified three genes, ADMATS1, TIE1, and BMP2, whose expression were significantly reduced in MXRA8-knockout tumors compared to control tumors. MXRA8 staining of a human breast cancer tissue array revealed higher levels of MXRA8 in primary tumors and metastases of aggressive tumor subtypes (TNBC and HER2+) compared to less aggressive, ER+ breast cancers. Our findings demonstrate for the first time that MXRA8 regulates the progression of human TNBC possibly through influencing the interaction of tumor cells with their microenvironment.
- Published
- 2023
- Full Text
- View/download PDF
21. L1CAM promotes ovarian cancer stemness and tumor initiation via FGFR1/SRC/STAT3 signaling
- Author
-
Marco Giordano, Alessandra Decio, Chiara Battistini, Micol Baronio, Fabrizio Bianchi, Alessandra Villa, Giovanni Bertalot, Stefano Freddi, Michela Lupia, Maria Giovanna Jodice, Paolo Ubezio, Nicoletta Colombo, Raffaella Giavazzi, and Ugo Cavallaro
- Subjects
L1CAM ,Ovarian cancer ,Cancer stem cells ,FGFR1 ,STAT3 ,Tumor initiation ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Cancer stem cells (CSC) have been implicated in tumor progression. In ovarian carcinoma (OC), CSC drive tumor formation, dissemination and recurrence, as well as drug resistance, thus contributing to the high death-to-incidence ratio of this disease. However, the molecular basis of such a pathogenic role of ovarian CSC (OCSC) has been elucidated only to a limited extent. In this context, the functional contribution of the L1 cell adhesion molecule (L1CAM) to OC stemness remains elusive. Methods The expression of L1CAM was investigated in patient-derived OCSC. The genetic manipulation of L1CAM in OC cells provided gain and loss-of-function models that were then employed in cell biological assays as well as in vivo tumorigenesis experiments to assess the role of L1CAM in OC cell stemness and in OCSC-driven tumor initiation. We applied antibody-mediated neutralization to investigate L1CAM druggability. Biochemical approaches were then combined with functional in vitro assays to study the molecular mechanisms underlying the functional role of L1CAM in OCSC. Results We report that L1CAM is upregulated in patient-derived OCSC. Functional studies showed that L1CAM promotes several stemness-related properties in OC cells, including sphere formation, tumor initiation and chemoresistance. These activities were repressed by an L1CAM-neutralizing antibody, pointing to L1CAM as a druggable target. Mechanistically, L1CAM interacted with and activated fibroblast growth factor receptor-1 (FGFR1), which in turn induced the SRC-mediated activation of STAT3. The inhibition of STAT3 prevented L1CAM-dependent OC stemness and tumor initiation. Conclusions Our study implicate L1CAM in the tumorigenic function of OCSC and point to the L1CAM/FGFR1/SRC/STAT3 signaling pathway as a novel driver of OC stemness. We also provide evidence that targeting this pathway can contribute to OC eradication.
- Published
- 2021
- Full Text
- View/download PDF
22. miRNA signaling networks in cancer stem cells
- Author
-
Kosuke Yoshida, Yusuke Yamamoto, and Takahiro Ochiya
- Subjects
microRNA ,Cancer stem cells (CSCs) ,Tumor initiation ,Therapeutic resistance ,Metastasis and recurrence ,Medicine (General) ,R5-920 ,Cytology ,QH573-671 - Abstract
Cancer stem cells (CSCs) are a small cell subpopulation in many cancer types and are involved in various processes of tumor progression, such as initiation, metastasis and recurrence. The distinguished features of CSCs include a variety of biological properties, including self-renewal, multidifferentiation, stemness marker expression, and resistance to chemotherapy and radiotherapy. Despite their great potential of clinical importance, the CSC signaling pathways are not well understood at the molecular level. MicroRNAs (miRNAs) are a class of endogenous noncoding RNAs that play an important role in the regulation of several cellular, physiological, and developmental processes. Aberrant miRNA expression is associated with many human diseases, including cancer. miRNAs have been implicated in the regulation of CSC properties; therefore, a better understanding of miRNA-induced modulation of CSC gene expression could aid in the identification of promising biomarkers and therapeutic targets. In the present review, we summarize the major findings of the impacts of miRNAs on CSC signaling networks; we then discuss the recent advances that have improved our understanding of CSC regulation by miRNA-mediated signaling networks and that may lead to the development of miRNA therapeutics specifically targeting CSCs.
- Published
- 2021
- Full Text
- View/download PDF
23. Long noncoding RNAs as tumorigenic factors and therapeutic targets for renal cell carcinoma
- Author
-
Haiyan Shen, Guomin Luo, and Qingjuan Chen
- Subjects
Long noncoding RNA ,Gene expression ,Renal cell carcinoma ,Tumor initiation ,Tumor progression ,Prognosis ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 ,Cytology ,QH573-671 - Abstract
Abstract Approximately 338,000 patients are diagnosed with kidney cancer worldwide each year, and renal cell carcinoma (RCC), which is derived from renal epithelium, accounts for more than ninety percent of the malignancy. Next generation RNA sequencing has enabled the identification of novel long noncoding RNAs (lncRNAs) in the past 10 years. Recent studies have provided extensive evidence that lncRNAs bind to chromatin modification proteins, transcription factors, RNA-binding proteins and microRNAs, and thereby modulate gene expression through regulating chromatin status, gene transcription, pre-mRNA splicing, mRNA decay and stability, protein translation and stability. In vitro and in vivo studies have demonstrated that over-expression of oncogenic lncRNAs and silencing of tumor suppressive lncRNAs are a common feature of human RCC, and that aberrant lncRNA expression is a marker for poor patient prognosis, and is essential for the initiation and progression of RCC. Because lncRNAs, compared with mRNAs, are expressed in a tissue-specific manner, aberrantly expressed lncRNAs can be better targeted for the treatment of RCC through screening small molecule compounds which block the interaction between lncRNAs and their binding proteins or microRNAs.
- Published
- 2021
- Full Text
- View/download PDF
24. On the Role of Glycolysis in Early Tumorigenesis—Permissive and Executioner Effects
- Author
-
Fabrizio Marcucci and Cristiano Rumio
- Subjects
glycolysis ,tumor initiation ,senescence ,oncoprotein ,DNA repair ,chromatin ,Cytology ,QH573-671 - Abstract
Reprogramming energy production from mitochondrial respiration to glycolysis is now considered a hallmark of cancer. When tumors grow beyond a certain size they give rise to changes in their microenvironment (e.g., hypoxia, mechanical stress) that are conducive to the upregulation of glycolysis. Over the years, however, it has become clear that glycolysis can also associate with the earliest steps of tumorigenesis. Thus, many of the oncoproteins most commonly involved in tumor initiation and progression upregulate glycolysis. Moreover, in recent years, considerable evidence has been reported suggesting that upregulated glycolysis itself, through its enzymes and/or metabolites, may play a causative role in tumorigenesis, either by acting itself as an oncogenic stimulus or by facilitating the appearance of oncogenic mutations. In fact, several changes induced by upregulated glycolysis have been shown to be involved in tumor initiation and early tumorigenesis: glycolysis-induced chromatin remodeling, inhibition of premature senescence and induction of proliferation, effects on DNA repair, O-linked N-acetylglucosamine modification of target proteins, antiapoptotic effects, induction of epithelial–mesenchymal transition or autophagy, and induction of angiogenesis. In this article we summarize the evidence that upregulated glycolysis is involved in tumor initiation and, in the following, we propose a mechanistic model aimed at explaining how upregulated glycolysis may play such a role.
- Published
- 2023
- Full Text
- View/download PDF
25. Cell Type-Specific Transcriptome Profiling Reveals a Role for Thioredoxin During Tumor Initiation.
- Author
-
Korte, Benjamin G., Giese, Morgan A., Ramakrishnan, Gayathri, Ma, Stella, Bennin, David, Rindy, Julie, Dewey, Colin N., and Huttenlocher, Anna
- Subjects
THIOREDOXIN ,EPITHELIAL cells ,REACTIVE oxygen species ,TRANSCRIPTOMES ,KERATINOCYTES - Abstract
Neutrophils in the tumor microenvironment exhibit altered functions. However, the changes in neutrophil behavior during tumor initiation remain unclear. Here we used Translating Ribosomal Affinity Purification (TRAP) and RNA sequencing to identify neutrophil, macrophage and transformed epithelial cell transcriptional changes induced by oncogenic Ras
G12V in larval zebrafish. We found that transformed epithelial cells and neutrophils, but not macrophages, had significant changes in gene expression in larval zebrafish. Interestingly, neutrophils had more significantly down-regulated genes, whereas gene expression was primarily upregulated in transformed epithelial cells. The antioxidant, thioredoxin (txn), a small thiol that regulates reduction-oxidation (redox) balance, was upregulated in transformed keratinocytes and neutrophils in response to oncogenic Ras. To determine the role of thioredoxin during tumor initiation, we generated a zebrafish thioredoxin mutant. We observed an increase in wound-induced reactive oxygen species signaling and neutrophil recruitment in thioredoxin-deficient zebrafish. Transformed keratinocytes also showed increased proliferation and reduced apoptosis in thioredoxin-deficient larvae. Using live imaging, we visualized neutrophil behavior near transformed cells and found increased neutrophil recruitment and altered motility dynamics. Finally, in the absence of neutrophils, transformed keratinocytes no longer exhibited increased proliferation in thioredoxin mutants. Taken together, our findings demonstrate that tumor initiation induces changes in neutrophil gene expression and behavior that can impact proliferation of transformed cells in the early tumor microenvironment. [ABSTRACT FROM AUTHOR]- Published
- 2022
- Full Text
- View/download PDF
26. CD44 as a tumor biomarker and therapeutic target
- Author
-
Hanxiao Xu, Mengke Niu, Xun Yuan, Kongming Wu, and Aiguo Liu
- Subjects
CD44 ,Cancer ,Cancer stem cells ,Epithelial-mesenchymal transition ,Tumor initiation ,Cancer progression ,Diseases of the blood and blood-forming organs ,RC633-647.5 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract CD44, a complex transmembrane glycoprotein, exists in multiple molecular forms, including the standard isoform CD44s and CD44 variant isoforms. CD44 participates in multiple physiological processes, and aberrant expression and dysregulation of CD44 contribute to tumor initiation and progression. CD44 represents a common biomarker of cancer stem cells, and promotes epithelial-mesenchymal transition. CD44 is involved in the regulation of diverse vital signaling pathways that modulate cancer proliferation, invasion, metastasis and therapy-resistance, and it is also modulated by a variety of molecules in cancer cells. In addition, CD44 can serve as an adverse prognostic marker among cancer population. The pleiotropic roles of CD44 in carcinoma potentially offering new molecular target for therapeutic intervention. Preclinical and clinical trials for evaluating the pharmacokinetics, efficacy and drug-related toxicity of CD44 monoclonal antibody have been carried out among tumors with CD44 expression. In this review, we focus on current data relevant to CD44, and outline CD44 structure, the regulation of CD44, functional properties of CD44 in carcinogenesis and cancer progression as well as the potential CD44-targeting therapy for cancer management.
- Published
- 2020
- Full Text
- View/download PDF
27. Cell Type-Specific Transcriptome Profiling Reveals a Role for Thioredoxin During Tumor Initiation
- Author
-
Benjamin G. Korte, Morgan A. Giese, Gayathri Ramakrishnan, Stella Ma, David Bennin, Julie Rindy, Colin N. Dewey, and Anna Huttenlocher
- Subjects
neutrophil ,migration ,tumor initiation ,thioredoxin (txn) ,gene expression ,keratinocyte ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Neutrophils in the tumor microenvironment exhibit altered functions. However, the changes in neutrophil behavior during tumor initiation remain unclear. Here we used Translating Ribosomal Affinity Purification (TRAP) and RNA sequencing to identify neutrophil, macrophage and transformed epithelial cell transcriptional changes induced by oncogenic RasG12V in larval zebrafish. We found that transformed epithelial cells and neutrophils, but not macrophages, had significant changes in gene expression in larval zebrafish. Interestingly, neutrophils had more significantly down-regulated genes, whereas gene expression was primarily upregulated in transformed epithelial cells. The antioxidant, thioredoxin (txn), a small thiol that regulates reduction-oxidation (redox) balance, was upregulated in transformed keratinocytes and neutrophils in response to oncogenic Ras. To determine the role of thioredoxin during tumor initiation, we generated a zebrafish thioredoxin mutant. We observed an increase in wound-induced reactive oxygen species signaling and neutrophil recruitment in thioredoxin-deficient zebrafish. Transformed keratinocytes also showed increased proliferation and reduced apoptosis in thioredoxin-deficient larvae. Using live imaging, we visualized neutrophil behavior near transformed cells and found increased neutrophil recruitment and altered motility dynamics. Finally, in the absence of neutrophils, transformed keratinocytes no longer exhibited increased proliferation in thioredoxin mutants. Taken together, our findings demonstrate that tumor initiation induces changes in neutrophil gene expression and behavior that can impact proliferation of transformed cells in the early tumor microenvironment.
- Published
- 2022
- Full Text
- View/download PDF
28. L1CAM promotes ovarian cancer stemness and tumor initiation via FGFR1/SRC/STAT3 signaling.
- Author
-
Giordano, Marco, Decio, Alessandra, Battistini, Chiara, Baronio, Micol, Bianchi, Fabrizio, Villa, Alessandra, Bertalot, Giovanni, Freddi, Stefano, Lupia, Michela, Jodice, Maria Giovanna, Ubezio, Paolo, Colombo, Nicoletta, Giavazzi, Raffaella, and Cavallaro, Ugo
- Subjects
- *
OVARIAN cancer , *CELL adhesion molecules , *CELLULAR signal transduction , *FIBROBLAST growth factors , *CANCER stem cells , *BIOLOGICAL assay - Abstract
Background: Cancer stem cells (CSC) have been implicated in tumor progression. In ovarian carcinoma (OC), CSC drive tumor formation, dissemination and recurrence, as well as drug resistance, thus contributing to the high death-to-incidence ratio of this disease. However, the molecular basis of such a pathogenic role of ovarian CSC (OCSC) has been elucidated only to a limited extent. In this context, the functional contribution of the L1 cell adhesion molecule (L1CAM) to OC stemness remains elusive. Methods: The expression of L1CAM was investigated in patient-derived OCSC. The genetic manipulation of L1CAM in OC cells provided gain and loss-of-function models that were then employed in cell biological assays as well as in vivo tumorigenesis experiments to assess the role of L1CAM in OC cell stemness and in OCSC-driven tumor initiation. We applied antibody-mediated neutralization to investigate L1CAM druggability. Biochemical approaches were then combined with functional in vitro assays to study the molecular mechanisms underlying the functional role of L1CAM in OCSC. Results: We report that L1CAM is upregulated in patient-derived OCSC. Functional studies showed that L1CAM promotes several stemness-related properties in OC cells, including sphere formation, tumor initiation and chemoresistance. These activities were repressed by an L1CAM-neutralizing antibody, pointing to L1CAM as a druggable target. Mechanistically, L1CAM interacted with and activated fibroblast growth factor receptor-1 (FGFR1), which in turn induced the SRC-mediated activation of STAT3. The inhibition of STAT3 prevented L1CAM-dependent OC stemness and tumor initiation. Conclusions: Our study implicate L1CAM in the tumorigenic function of OCSC and point to the L1CAM/FGFR1/SRC/STAT3 signaling pathway as a novel driver of OC stemness. We also provide evidence that targeting this pathway can contribute to OC eradication. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
29. The Role of ARID1A in Tumors: Tumor Initiation or Tumor Suppression?
- Author
-
Xu, Shouying and Tang, Chao
- Subjects
CELLULAR signal transduction ,CELL physiology ,CHROMATIN ,TUMORS ,CANCER invasiveness - Abstract
Genes encoding subunits of SWItch/Sucrose Non-Fermenting (SWI/SNF) chromatin remodeling complexes are collectively mutated in 20% of all human cancers, among which the AT-rich interacting domain−containing protein 1A (ARID1A , also known as BAF250a , B120 , C1orf4 , Osa1) that encodes protein ARID1A is the most frequently mutated, and mutations in ARID1A have been found in various types of cancer. ARID1A is thought to play a significant role both in tumor initiation and in tumor suppression, which is highly dependent upon context. Recent molecular mechanistic research has revealed that ARID1A participates in tumor progression through its effects on control of cell cycle, modulation of cellular functions such as EMT, and regulation of various signaling pathways. In this review, we synthesize a mechanistic understanding of the role of ARID1A in human tumor initiation as well as in tumor suppression and further discuss the implications of these new discoveries for potential cancer intervention. We also highlight the mechanisms by which mutations affecting the subunits in SWI/SNF complexes promote cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
30. High Variability in Cellular Proliferation, Gene Expression, and Cytokine Production in the Nonneoplastic Colonic Epithelium of Young Apc+/Min-FCCC Mice.
- Author
-
Leystra, Alyssa A., Harvey, Kristen N., Kaunga, Esther, Hensley, Harvey, Vanderveer, Lisa A., Devarajan, Karthik, and Clapper, Margie L.
- Subjects
CYTOKINES ,CELL proliferation ,GENE expression ,COLORECTAL cancer ,EPITHELIUM - Abstract
An urgent need exists to identify efficacious therapeutic preventive interventions for individuals who are at high risk of developing colorectal cancer. To maximize the benefits of preventive intervention, it is vital to identify the time interval during which the initiation of a preventive intervention will lead to an optimal outcome. The goal of the present study was to determine if oncogenic events can be detected in the nonneoplastic colonic mucosa of Apc
+/Min-FCCC mice prior to formation of the first adenoma, thus defining an earlier point of intervention along the cancer continuum. Tissues taken at three potential points of intervention were characterized: prior to Apc mutation (wild type Apc+/+-FCCC mice); after initiation but prior to colon adenoma formation (tumor-free Apc+/Min-FCCC mice); and after formation of the first colon adenoma (tumor-bearing Apc+/Min-FCCC mice). Experimentation focused on molecular processes that are dysregulated in early colon lesions: 1) cellular proliferation (proliferative index and size of the proliferative zone); 2) cellular stemness (expression of Ascl2 , Grem1 , Lgr5 and Muc2); 3) EGFR signaling (expression of Ereg); and 4) inflammation (expression of Mmp9 , Ptsg2 , and Reg4 , as well as secretion of 18 cytokines involved in immune activation and response). Interestingly, the nonneoplastic colonic mucosa of wild type, tumor-free Apc+/Min-FCCC , and tumor-bearing Apc+/Min-FCCC mice did not display significant differences in average epithelial cell proliferation (fold change 0.8–1.3, p≥0.11), mucosal gene expression (fold change 0.8–1.4, p≥0.22), or secretion of specific cytokines from colonic mucosa (fold change 0.2–1.5, p≥0.06). However, the level of cytokine secretion was highly variable, with many (22% of wild type, 31% of tumor-free Apc+/Min-FCCC , and 31% of tumor-bearing Apc+/Min-FCCC ) mice categorized as outliers (> 1.5 x interquartile ranges below the first quartile or above the third quartile) due to elevated expression of at least one cytokine. In summary, no differences were observed in proliferation, stemness, and EGFR signaling in the colonic mucosa of wild type vs Apc+/Min-FCCC mice, with low baseline cytokine expression, prior to the formation of the first colon adenoma. The results of this study provide valuable baseline data to inform the design of future cancer prevention studies. [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
31. Comparative lineage tracing reveals cellular preferences for prostate cancer initiation
- Author
-
Wang, Zhu A and Shen, Michael M
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Stem Cell Research ,Urologic Diseases ,Cancer ,Prostate Cancer ,Aging ,2.1 Biological and endogenous factors ,Aetiology ,cell of origin ,mouse models ,prostate cancer ,tumor initiation ,Medical biochemistry and metabolomics ,Oncology and carcinogenesis - Abstract
The interplay of different cell types of origin and distinct oncogenic mutations may determine the tumor subtype. We have recently found that although both basal and luminal epithelial cells can initiate prostate tumorigenesis, the latter are more likely to undergo transformation in response to a range of oncogenic events.
- Published
- 2015
32. The Role of ARID1A in Tumors: Tumor Initiation or Tumor Suppression?
- Author
-
Shouying Xu and Chao Tang
- Subjects
ARID1A ,SWI/SNF ,tumor initiation ,tumor suppression ,synthetic lethality ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Genes encoding subunits of SWItch/Sucrose Non-Fermenting (SWI/SNF) chromatin remodeling complexes are collectively mutated in 20% of all human cancers, among which the AT-rich interacting domain−containing protein 1A (ARID1A, also known as BAF250a, B120, C1orf4, Osa1) that encodes protein ARID1A is the most frequently mutated, and mutations in ARID1A have been found in various types of cancer. ARID1A is thought to play a significant role both in tumor initiation and in tumor suppression, which is highly dependent upon context. Recent molecular mechanistic research has revealed that ARID1A participates in tumor progression through its effects on control of cell cycle, modulation of cellular functions such as EMT, and regulation of various signaling pathways. In this review, we synthesize a mechanistic understanding of the role of ARID1A in human tumor initiation as well as in tumor suppression and further discuss the implications of these new discoveries for potential cancer intervention. We also highlight the mechanisms by which mutations affecting the subunits in SWI/SNF complexes promote cancer.
- Published
- 2021
- Full Text
- View/download PDF
33. High Variability in Cellular Proliferation, Gene Expression, and Cytokine Production in the Nonneoplastic Colonic Epithelium of Young Apc+/Min-FCCC Mice
- Author
-
Alyssa A. Leystra, Kristen N. Harvey, Esther Kaunga, Harvey Hensley, Lisa A. Vanderveer, Karthik Devarajan, and Margie L. Clapper
- Subjects
colorectal cancer prevention ,cancer prevention ,cytokine signaling ,tumor initiation ,chemoprevention ,immunoprevention ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
An urgent need exists to identify efficacious therapeutic preventive interventions for individuals who are at high risk of developing colorectal cancer. To maximize the benefits of preventive intervention, it is vital to identify the time interval during which the initiation of a preventive intervention will lead to an optimal outcome. The goal of the present study was to determine if oncogenic events can be detected in the nonneoplastic colonic mucosa of Apc+/Min-FCCC mice prior to formation of the first adenoma, thus defining an earlier point of intervention along the cancer continuum. Tissues taken at three potential points of intervention were characterized: prior to Apc mutation (wild type Apc+/+-FCCC mice); after initiation but prior to colon adenoma formation (tumor-free Apc+/Min-FCCC mice); and after formation of the first colon adenoma (tumor-bearing Apc+/Min-FCCC mice). Experimentation focused on molecular processes that are dysregulated in early colon lesions: 1) cellular proliferation (proliferative index and size of the proliferative zone); 2) cellular stemness (expression of Ascl2, Grem1, Lgr5 and Muc2); 3) EGFR signaling (expression of Ereg); and 4) inflammation (expression of Mmp9, Ptsg2, and Reg4, as well as secretion of 18 cytokines involved in immune activation and response). Interestingly, the nonneoplastic colonic mucosa of wild type, tumor-free Apc+/Min-FCCC, and tumor-bearing Apc+/Min-FCCC mice did not display significant differences in average epithelial cell proliferation (fold change 0.8–1.3, p≥0.11), mucosal gene expression (fold change 0.8–1.4, p≥0.22), or secretion of specific cytokines from colonic mucosa (fold change 0.2–1.5, p≥0.06). However, the level of cytokine secretion was highly variable, with many (22% of wild type, 31% of tumor-free Apc+/Min-FCCC, and 31% of tumor-bearing Apc+/Min-FCCC) mice categorized as outliers (> 1.5 x interquartile ranges below the first quartile or above the third quartile) due to elevated expression of at least one cytokine. In summary, no differences were observed in proliferation, stemness, and EGFR signaling in the colonic mucosa of wild type vs Apc+/Min-FCCC mice, with low baseline cytokine expression, prior to the formation of the first colon adenoma. The results of this study provide valuable baseline data to inform the design of future cancer prevention studies.
- Published
- 2021
- Full Text
- View/download PDF
34. Signaling levels mold the RAS mutation tropism of urethane
- Author
-
Siqi Li and Christopher M Counter
- Subjects
protooncogenes ,RAS ,oncogenesis ,codon bias ,tumor initiation ,carcinogenesis ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
RAS genes are commonly mutated in human cancer. Despite many possible mutations, individual cancer types often have a ‘tropism’ towards a specific subset of RAS mutations. As driver mutations, these patterns ostensibly originate from normal cells. High oncogenic RAS activity causes oncogenic stress and different oncogenic mutations can impart different levels of activity, suggesting a relationship between oncoprotein activity and RAS mutation tropism. Here, we show that changing rare codons to common in the murine Kras gene to increase protein expression shifts tumors induced by the carcinogen urethane from arising from canonical Q61 to biochemically less active G12 Kras driver mutations, despite the carcinogen still being biased towards generating Q61 mutations. Conversely, inactivating the tumor suppressor p53 to blunt oncogenic stress partially reversed this effect, restoring Q61 mutations. One interpretation of these findings is that the RAS mutation tropism of urethane arises from selection in normal cells for specific mutations that impart a narrow window of signaling that promotes proliferation without causing oncogenic stress.
- Published
- 2021
- Full Text
- View/download PDF
35. Danger zone
- Author
-
Zohra Butt and Ian Prior
- Subjects
tumour initiation ,RAS ,oncogenesis ,codon bias ,tumor initiation ,carcinogenesis ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
What level of Ras genes activity leads to the development of cancer?
- Published
- 2021
- Full Text
- View/download PDF
36. Long noncoding RNAs as tumorigenic factors and therapeutic targets for renal cell carcinoma.
- Author
-
Shen, Haiyan, Luo, Guomin, and Chen, Qingjuan
- Subjects
- *
LINCRNA , *RENAL cell carcinoma , *SMALL molecules , *NUCLEOTIDE sequence , *RNA-binding proteins , *SYNCRIP protein , *INTRONS - Abstract
Approximately 338,000 patients are diagnosed with kidney cancer worldwide each year, and renal cell carcinoma (RCC), which is derived from renal epithelium, accounts for more than ninety percent of the malignancy. Next generation RNA sequencing has enabled the identification of novel long noncoding RNAs (lncRNAs) in the past 10 years. Recent studies have provided extensive evidence that lncRNAs bind to chromatin modification proteins, transcription factors, RNA-binding proteins and microRNAs, and thereby modulate gene expression through regulating chromatin status, gene transcription, pre-mRNA splicing, mRNA decay and stability, protein translation and stability. In vitro and in vivo studies have demonstrated that over-expression of oncogenic lncRNAs and silencing of tumor suppressive lncRNAs are a common feature of human RCC, and that aberrant lncRNA expression is a marker for poor patient prognosis, and is essential for the initiation and progression of RCC. Because lncRNAs, compared with mRNAs, are expressed in a tissue-specific manner, aberrantly expressed lncRNAs can be better targeted for the treatment of RCC through screening small molecule compounds which block the interaction between lncRNAs and their binding proteins or microRNAs. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
37. Fully Automatic Calibration of Tumor-Growth Models Using a Single mpMRI Scan.
- Author
-
Scheufele, Klaudius, Subramanian, Shashank, and Biros, George
- Subjects
- *
TUMOR growth , *PARTIAL differential equations , *MAGNETIC resonance imaging , *TUMOR markers , *CALIBRATION , *CELL migration , *DECOMPOSITION method - Abstract
Our objective is the calibration of mathematical tumor growth models from a single multiparametric scan. The target problem is the analysis of preoperative Glioblastoma (GBM) scans. To this end, we present a fully automatic tumor-growth calibration methodology that integrates a single-species reaction-diffusion partial differential equation (PDE) model for tumor progression with multiparametric Magnetic Resonance Imaging (mpMRI) scans to robustly extract patient specific biomarkers i.e., estimates for (i) the tumor cell proliferation rate, (ii) the tumor cell migration rate, and (iii) the original, localized site(s) of tumor initiation. Our method is based on a sparse reconstruction algorithm for the tumor initial location (TIL). This problem is particularly challenging due to nonlinearity, ill-posedeness, and ill conditioning. We propose a coarse-to-fine multi-resolution continuation scheme with parameter decomposition to stabilize the inversion. We demonstrate robustness and practicality of our method by applying the proposed method to clinical data of 206 GBM patients. We analyze the extracted biomarkers and relate tumor origin with patient overall survival by mapping the former into a common atlas space. We present preliminary results that suggest improved accuracy for prediction of patient overall survival when a set of imaging features is augmented with estimated biophysical parameters. All extracted features, tumor initial positions, and biophysical growth parameters are made publicly available for further analysis. To our knowledge, this is the first fully automatic scheme that can handle multifocal tumors and can localize the TIL to a few millimeters. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
38. CD44 as a tumor biomarker and therapeutic target.
- Author
-
Xu, Hanxiao, Niu, Mengke, Yuan, Xun, Wu, Kongming, and Liu, Aiguo
- Subjects
- *
BIOMARKERS , *EPITHELIAL-mesenchymal transition , *CANCER stem cells , *HUMAN carcinogenesis , *CANCER invasiveness , *TUMOR markers - Abstract
CD44, a complex transmembrane glycoprotein, exists in multiple molecular forms, including the standard isoform CD44s and CD44 variant isoforms. CD44 participates in multiple physiological processes, and aberrant expression and dysregulation of CD44 contribute to tumor initiation and progression. CD44 represents a common biomarker of cancer stem cells, and promotes epithelial-mesenchymal transition. CD44 is involved in the regulation of diverse vital signaling pathways that modulate cancer proliferation, invasion, metastasis and therapy-resistance, and it is also modulated by a variety of molecules in cancer cells. In addition, CD44 can serve as an adverse prognostic marker among cancer population. The pleiotropic roles of CD44 in carcinoma potentially offering new molecular target for therapeutic intervention. Preclinical and clinical trials for evaluating the pharmacokinetics, efficacy and drug-related toxicity of CD44 monoclonal antibody have been carried out among tumors with CD44 expression. In this review, we focus on current data relevant to CD44, and outline CD44 structure, the regulation of CD44, functional properties of CD44 in carcinogenesis and cancer progression as well as the potential CD44-targeting therapy for cancer management. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
39. Identification and Characterization of Tumor-Initiating Cells in Multiple Myeloma.
- Author
-
Gao, Minjie, Bai, Hua, Jethava, Yogesh, Wu, Yujie, Zhu, Yuqi, Yang, Ye, Xia, Jiliang, Cao, Huojun, Franqui-Machin, Reinaldo, Nadiminti, Kalyan, Thomas, Gregory S, Salama, Mohamed E, Altevogt, Peter, Bishop, Gail, Tomasson, Michael, Janz, Siegfried, Shi, Jumei, Chen, Lijuan, Frech, Ivana, and Tricot, Guido
- Subjects
- *
REVERSE transcriptase polymerase chain reaction , *MULTIPLE myeloma , *INDUCED pluripotent stem cells , *FLUORESCENT antibody technique , *RESEARCH , *XENOGRAFTS , *ANIMAL experimentation , *CARCINOGENESIS , *RESEARCH methodology , *EVALUATION research , *MEDICAL cooperation , *COMPARATIVE studies , *STEM cells , *RESEARCH funding , *MICE , *DRUG resistance in cancer cells - Abstract
Background: Treatment failures in cancers, including multiple myeloma (MM), are most likely due to the persistence of a minor population of tumor-initiating cells (TICs), which are noncycling or slowly cycling and very drug resistant.Methods: Gene expression profiling and real-time quantitative reverse transcription polymerase chain reaction were employed to define genes differentially expressed between the side-population cells, which contain the TICs, and the main population of MM cells derived from 11 MM patient samples. Self-renewal potential was analyzed by clonogenicity and drug resistance of CD24+ MM cells. Flow cytometry (n = 60) and immunofluorescence (n = 66) were applied on MM patient samples to determine CD24 expression. Therapeutic effects of CD24 antibodies were tested in xenograft MM mouse models containing three to six mice per group.Results: CD24 was highly expressed in the side-population cells, and CD24+ MM cells exhibited high expression of induced pluripotent or embryonic stem cell genes. CD24+ MM cells showed increased clonogenicity, drug resistance, and tumorigenicity. Only 10 CD24+ MM cells were required to develop plasmacytomas in mice (n = three of five mice after 27 days). The frequency of CD24+ MM cells was highly variable in primary MM samples, but the average of CD24+ MM cells was 8.3% after chemotherapy and in complete-remission MM samples with persistent minimal residual disease compared with 1.0% CD24+ MM cells in newly diagnosed MM samples (n = 26). MM patients with a high initial percentage of CD24+ MM cells had inferior progression-free survival (hazard ratio [HR] = 3.81, 95% confidence interval [CI] = 5.66 to 18.34, P < .001) and overall survival (HR = 3.87, 95% CI = 16.61 to 34.39, P = .002). A CD24 antibody inhibited MM cell growth and prevented tumor progression in vivo.Conclusion: Our studies demonstrate that CD24+ MM cells maintain the TIC features of self-renewal and drug resistance and provide a target for myeloma therapy. [ABSTRACT FROM AUTHOR]- Published
- 2020
- Full Text
- View/download PDF
40. Dichotomous roles of claudins as tumor promoters or suppressors: lessons from knockout mice.
- Author
-
Kage, Hidenori, Flodby, Per, Zhou, Beiyun, and Borok, Zea
- Subjects
- *
TIGHT junctions , *KNOCKOUT mice , *COCARCINOGENS , *ANTIBODY-dependent cell cytotoxicity , *MONOCLONAL antibodies , *CHIMERIC proteins , *CLAUDINS - Abstract
Claudins are a family of integral tight junction proteins that regulate paracellular permeability in polarized epithelia. Overexpression or reduction of claudins can both promote and limit cancer progression, revealing complex dichotomous roles for claudins depending on cellular context. In contrast, recent studies demonstrating tumor formation in claudin knockout mouse models indicate a role for several claudin family members in suppressing tumor initiation. For example, intestine-specific claudin-7 knockout mice spontaneously develop atypical hyperplasia and intestinal adenomas, while claudin-18 knockout mice develop carcinomas in the lung and stomach. Claudin-4, -11, and -15 knockout mice show increased cell proliferation and/or hyperplasia in urothelium, Sertoli cells, and small intestinal crypts, respectively, possibly a precursor to cancer development. Pathways implicated in both cell proliferation and tumorigenesis include Yap/Taz and insulin-like growth factor-1 receptor (IGF-1R)/Akt pathways, among others. Consistent with the tumor suppressive role of claudins shown in mice, in humans, claudin-low breast cancer has been described as a distinct entity with a poor prognosis, and claudin-18-Rho GTPase activating protein 26 (CLDN18-ARHGAP26) fusion protein as a driver gene aberration in diffuse-type gastric cancer due to effects on RhoA. Paradoxically, claudins have also garnered interest as targets for therapy, as they are sometimes aberrantly expressed in cancer cells, which may or may not promote cancer progression. For example, a chimeric monoclonal antibody which targets cells expressing claudin-18.2 through antibody-dependent cell-mediated cytotoxicity has shown promise in multiple phase II studies. In this review, we focus on new findings supporting a tumor suppressive role for claudins during cancer initiation. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
41. 7.342 Cancer Biology: From Basic Research to the Clinic, Fall 2004
- Author
-
Kim, Carla, Haigis, Kevin, Kim, Carla, and Haigis, Kevin
- Abstract
This course is one of many Advanced Undergraduate Seminars offered by the Biology Department at MIT. These seminars are tailored for students with an interest in using primary research literature to discuss and learn about current biological research in a highly interactive setting. In 1971, President Nixon declared the "War on Cancer," but after three decades the war is still raging. How much progress have we made toward winning the war and what are we doing to improve the fight? Understanding the molecular and cellular events involved in tumor formation, progression, and metastasis is crucial to the development of innovative therapy for cancer patients. Insights into these processes have been gleaned through basic research using biochemical, molecular, and genetic analysis in yeast, C. elegans, mice, and cell culture models. We will explore the laboratory tools and techniques used to perform cancer research, major discoveries in cancer biology, and the medical implications of these breakthroughs. A focus of the class will be critical analysis of the primary literature to foster understanding of the strengths and limitations of various approaches to cancer research. Special attention will be made to the clinical implications of cancer research performed in model organisms and the prospects for ending the battle with this devastating disease.
- Published
- 2023
42. Prospective Isolation and Comparison of Human Germinal Matrix and Glioblastoma EGFR+ Populations with Stem Cell Properties
- Author
-
Jessica Tome-Garcia, Rut Tejero, German Nudelman, Raymund L. Yong, Robert Sebra, Huaien Wang, Mary Fowkes, Margret Magid, Martin Walsh, Violeta Silva-Vargas, Elena Zaslavsky, Roland H. Friedel, Fiona Doetsch, and Nadejda M. Tsankova
- Subjects
neural stem cells ,glioma stem cells ,germinal matrix ,glioblastoma ,tumor initiation ,neurosphere ,FACS ,RNA-seq ,transcriptome ,EGFR ,Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Characterization of non-neoplastic and malignant human stem cell populations in their native state can provide new insights into gliomagenesis. Here we developed a purification strategy to directly isolate EGFR+/− populations from human germinal matrix (GM) and adult subventricular zone autopsy tissues, and from de novo glioblastoma (GBM) resections, enriching for cells capable of binding EGF ligand (LBEGFR+), and uniquely compared their functional and molecular properties. LBEGFR+ populations in both GM and GBM encompassed all sphere-forming cells and displayed proliferative stem cell properties in vitro. In xenografts, LBEGFR+ GBM cells showed robust tumor initiation and progression to high-grade, infiltrative gliomas. Whole-transcriptome sequencing analysis confirmed enrichment of proliferative pathways in both developing and neoplastic freshly isolated EGFR+ populations, and identified both unique and shared sets of genes. The ability to prospectively isolate stem cell populations using native ligand-binding capacity opens new doors onto understanding both normal human development and tumor cell biology.
- Published
- 2017
- Full Text
- View/download PDF
43. Chemotherapy-Induced Ca2+ Release Stimulates Breast Cancer Stem Cell Enrichment
- Author
-
Haiquan Lu, Ivan Chen, Larissa A. Shimoda, Youngrok Park, Chuanzhao Zhang, Linh Tran, Huimin Zhang, and Gregg L. Semenza
- Subjects
breast cancer stem cell ,chemotherapy ,hypoxia-inducible factors ,pluripotency factors ,tumor initiation ,metastasis ,tumor recurrence ,Biology (General) ,QH301-705.5 - Abstract
Breast cancer stem cells (BCSCs) play a critical role in tumor recurrence and metastasis. Exposure of breast cancer cells to chemotherapy leads to an enrichment of BCSCs. Here, we find that chemotherapy induces the expression of glutathione S-transferase omega 1 (GSTO1), which is dependent on hypoxia-inducible factor 1 (HIF-1) and HIF-2. Knockdown of GSTO1 expression abrogates carboplatin-induced BCSC enrichment, decreases tumor initiation and metastatic capacity, and delays tumor recurrence after chemotherapy. GSTO1 interacts with the ryanodine receptor RYR1 and promotes calcium release from the endoplasmic reticulum. Increased cytosolic calcium levels activate PYK2 → SRC → STAT3 signaling, leading to increased expression of pluripotency factors and BCSC enrichment. HIF inhibition blocks chemotherapy-induced GSTO1 expression and BCSC enrichment. Combining HIF inhibitors with chemotherapy may improve clinical outcome in breast cancer.
- Published
- 2017
- Full Text
- View/download PDF
44. Dissecting the Dual Nature of Hyaluronan in the Tumor Microenvironment
- Author
-
Muhan Liu, Cornelia Tolg, and Eva Turley
- Subjects
hyaluronan ,hyaluronan receptors ,tumor microenvironment ,cancer resistance ,tumor initiation ,CD44 ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Hyaluronan (HA) is a glycosaminoglycan with a simple structure but diverse and often opposing functions. The biological activities of this polysaccharide depend on its molecular weight and the identity of interacting receptors. HA is initially synthesized as high molecular-weight (HMW) polymers, which maintain homeostasis and restrain cell proliferation and migration in normal tissues. These HMW-HA functions are mediated by constitutively expressed receptors including CD44, LYVE-1, and STABILIN2. During normal processes such as tissue remodeling and wound healing, HMW-HA is fragmented into low molecular weight polymers (LMW-HA) by hyaluronidases and free radicals, which promote inflammation, immune cell recruitment and the epithelial cell migration. These functions are mediated by RHAMM and TLR2,4, which coordinate signaling with CD44 and other HA receptors. Tumor cells hijack the normally tightly regulated HA production/fragmentation associated with wound repair/remodeling, and these HA functions participate in driving and maintaining malignant progression. However, elevated HMW-HA production in the absence of fragmentation is linked to cancer resistance. The controlled production of HA polymer sizes and their functions are predicted to be key to dissecting the role of microenvironment in permitting or restraining the oncogenic potential of tissues. This review focuses on the dual nature of HA in cancer initiation vs. resistance, and the therapeutic potential of HA for chemo-prevention and as a target for cancer management.
- Published
- 2019
- Full Text
- View/download PDF
45. The microRNA target site profile is a novel biomarker in the immunotherapy response.
- Author
-
Bai Y, Li Y, Qin Y, Yang X, Tseng GC, Kim S, and Park HJ
- Abstract
MicroRNAs (miRNAs) bind on the 3' untranslated region (3'UTR) of messenger RNAs (mRNAs) and regulate mRNA expression in physiological and pathological conditions, including cancer. Thus, studies have identified miRNAs as potential biomarkers by correlating the miRNA expression with the expression of important mRNAs and/or clinical outcomes in cancers. However, tumors undergo pervasive 3'UTR shortening/lengthening events through alternative polyadenylation (APA), which varies the number of miRNA target sites in mRNA, raising the number of miRNA target sites (numTS) as another important regulatory axis of the miRNA binding effects. In this study, we developed the first statistical method, BIOMATA-APA, to identify predictive miRNAs based on numTS features. Running BIOMATA-APA on The Cancer Genome Atlas (TCGA) and independent cohort data both with immunotherapy and no immunotherapy, we demonstrated for the first time that the numTS feature 1) distinguishes different cancer types, 2) predicts tumor proliferation and immune infiltration status, 3) explains more variation in the proportion of tumor-infiltrating immune cells, 4) predicts response to immune checkpoint blockade (ICB) therapy, and 5) adds prognostic power beyond clinical and miRNA expression. To the best of our knowledge, this is the first pan-cancer study to systematically demonstrate numTS as a novel type of biomarker representing the miRNA binding effects underlying tumorigenesis and pave the way to incorporate miRNA target sites for miRNA biomarker identification. Another advantage of examining the miRNA binding effect using numTS is that it requires only RNA-Seq data, not miRNAs, thus resulting in high power in the miRNA biomarker identification., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Bai, Li, Qin, Yang, Tseng, Kim and Park.)
- Published
- 2023
- Full Text
- View/download PDF
46. A systematic review of preclinical studies evaluating the association between nicotine and the initiation and progression of cancer.
- Author
-
Kim MM, Steffensen I, Miguel RTD, Babic T, Johnson AD, Potts R, and Junker CS
- Abstract
Background: The association between cigarette smoking and the increased risk of many cancers is well established. Conversely, epidemiological studies of smokeless tobacco demonstrate decreased risk, or no elevated risk, of certain cancers versus smoking. However, it is unclear what role, if any, nicotine plays in these associations. The objective of this systematic review was to synthesize the available evidence from preclinical studies that examined the potential association between nicotine and the initiation and/or progression of cancer., Methods: MEDLINE, Embase, PsychInfo, and Cochrane Database of Systematic Reviews were searched for articles published from inception until February 13, 2022. Studies were eligible for inclusion if they evaluated animal cancer or tumor models, compared nicotine and non-nicotine groups, and evaluated measures of cancer initiation or progression., Results: Among 1,137 identified articles, 61 were included in qualitative synthesis. Twelve studies reported data on tumor initiation, and 54 studies reported data on tumor progression. The majority of the tumor initiation studies did not identify an association between nicotine exposure and an increased risk of spontaneous tumor initiation. Results of tumor progression studies were inconsistent and varied across the reported measures, cancer type being evaluated, and animal cancer model used. Overall, the quality of reporting was poor, with many studies not demonstrating a high level of internal and/or external validity., Conclusions: In conclusion, although animal models have provided invaluable data for human health risk assessments of chemical exposures, the heterogeneity across the studies included in this systematic review make the interpretation and generalizability of the results difficult., Competing Interests: Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://atm.amegroups.com/article/view/10.21037/atm-23-1710/coif). M.M.K. serviced as a full-time employee of Reynolds American until March 31, 2023 and received restricted company shares as part of the company benefits package. In April of 2023, M.M.K. began a new full-time position at Thera-Business. Thera-Business Inc. has provided regulatory science consulting services to RAI Services Company, a wholly-owned subsidiary of British American Tobacco. I.S. is an employee of Thera-Business. Thera-Business Inc. has provided consulting services to RAI Services Company, a wholly-owned subsidiary of British American Tobacco. R.T.D.M. is an employee of Thera-Business. Thera-Business Inc. has provided consulting services to RAI Services Company, a wholly-owned subsidiary of British American Tobacco. A.D.J. is a full-time employee of Reynolds American. R.P. is a full-time employee of Reynolds American and receives restricted shares as a part of the benefits package. Reynolds American is a sponsor that has paid Thera-Business for contracted regulatory research services. C.S.J. is a full-time employee of Reynolds American, and receive restricted shares as a part of the benefits package. Reynolds American is a sponsor that has paid Thera-Business for contracted regulatory research services. T.B. has no conflicts of interest to declare., (2023 Annals of Translational Medicine. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
47. The Pros and Cons of Incorporating Transcriptomics in the Age of Precision Oncology.
- Author
-
Lin, Victor T G and Yang, Eddy S
- Subjects
- *
NON-small-cell lung carcinoma , *ONCOLOGY - Abstract
The treatment of cancer continues to evolve toward personalized therapies based on individual patient and tumor characteristics. Our successes and failures in adopting a precision-oncology approach have demonstrated the utmost importance in identifying the proper predictive biomarkers of response. Until recently, most biomarkers were identified using immunohistochemistry for protein expression or single-gene analysis to identify targetable alterations. With the rapid propagation of next-generation sequencing to evaluate tumor tissue and "liquid biopsies," identification of genomic biomarkers is now standard, particularly in non-small cell lung cancer, for which there is now an extensive catalog of biomarker-directed therapies with more anticipated to come. Despite these great strides, it has also become apparent that using genomic biomarkers alone will be insufficient, as it has been consistently shown that at least one-half of patients who undergo tumor genomic profiling have no actionable alteration. This is perhaps to be expected given the remarkable breadth of nongenetic factors that contribute to tumor initiation and progression. Some have proposed that the next logical step is to use transcriptome profiling to define new biomarkers of response to targeted agents. Recently, results from the WINTHER trial were published, specifically investigating the use of transcriptomics to improve match rates over genomic next-generation sequencing alone. In this review, we discuss the complexities of precision-oncology efforts and appraise the available evidence supporting the incorporation of transcriptomic data into the precision-oncology framework in the historical context of the development of biomarkers for directing cancer therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
48. Dissecting the Dual Nature of Hyaluronan in the Tumor Microenvironment.
- Author
-
Liu, Muhan, Tolg, Cornelia, and Turley, Eva
- Subjects
HYALURONIC acid ,TUMOR microenvironment ,GLYCOSAMINOGLYCANS ,POLYSACCHARIDES ,HOMEOSTASIS ,CELL proliferation - Abstract
Hyaluronan (HA) is a glycosaminoglycan with a simple structure but diverse and often opposing functions. The biological activities of this polysaccharide depend on its molecular weight and the identity of interacting receptors. HA is initially synthesized as high molecular-weight (HMW) polymers, which maintain homeostasis and restrain cell proliferation and migration in normal tissues. These HMW-HA functions are mediated by constitutively expressed receptors including CD44, LYVE-1, and STABILIN2. During normal processes such as tissue remodeling and wound healing, HMW-HA is fragmented into low molecular weight polymers (LMW-HA) by hyaluronidases and free radicals, which promote inflammation, immune cell recruitment and the epithelial cell migration. These functions are mediated by RHAMM and TLR2,4, which coordinate signaling with CD44 and other HA receptors. Tumor cells hijack the normally tightly regulated HA production/fragmentation associated with wound repair/remodeling, and these HA functions participate in driving and maintaining malignant progression. However, elevated HMW-HA production in the absence of fragmentation is linked to cancer resistance. The controlled production of HA polymer sizes and their functions are predicted to be key to dissecting the role of microenvironment in permitting or restraining the oncogenic potential of tissues. This review focuses on the dual nature of HA in cancer initiation vs. resistance, and the therapeutic potential of HA for chemo-prevention and as a target for cancer management. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
49. circCTIC1 promotes the self-renewal of colon TICs through BPTF-dependent c-Myc expression.
- Author
-
Zhan, Wei, Liao, Xin, Wang, Yuan, Li, Lianghe, Li, Jin, Chen, Zhongsheng, Tian, Tian, and He, Jingdong
- Subjects
- *
FIBROBLAST growth factor 2 , *CIRCULAR RNA - Published
- 2019
- Full Text
- View/download PDF
50. Novel therapies hijack the blood–brain barrier to eradicate glioblastoma cancer stem cells.
- Author
-
Vengoji, Raghupathy, Ponnusamy, Moorthy P, Rachagani, Satyanarayana, Mahapatra, Sidharth, Batra, Surinder K, Shonka, Nicole, and Macha, Muzafar A
- Subjects
- *
CANCER stem cells , *BLOOD-brain barrier , *RADIOTHERAPY - Abstract
Glioblastoma (GBM) is amongst the most aggressive brain tumors with a dismal prognosis. Despite significant advances in the current multimodality therapy including surgery, postoperative radiotherapy (RT) and temozolomide (TMZ)-based concomitant and adjuvant chemotherapy (CT), tumor recurrence is nearly universal with poor patient outcomes. These limitations are in part due to poor drug penetration through the blood–brain barrier (BBB) and resistance to CT and RT by a small population of cancer cells recognized as tumor-initiating cells or cancer stem cells (CSCs). Though CT and RT kill the bulk of the tumor cells, they fail to affect CSCs, resulting in their enrichment and their development into more refractory tumors. Therefore, identifying the mechanisms of resistance and developing therapies that specifically target CSCs can improve response, prevent the development of refractory tumors and increase overall survival of GBM patients. Small molecule inhibitors that can breach the BBB and selectively target CSCs are emerging. In this review, we have summarized the recent advancements in understanding the GBM CSC-specific signaling pathways, the CSC–tumor microenvironment niche that contributes to CT and RT resistance and the use of novel combination therapies of small molecule inhibitors that may be used in conjunction with TMZ-based chemoradiation for effective management of GBM. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.