Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Cancer Immunol Res ; 12(3): 287-295, 2024 03 04.
Article in English | MEDLINE | ID: mdl-38345376

ABSTRACT

Immune checkpoint blockade (ICB) can induce durable cancer remission. However, only a small subset of patients gains benefits. While tumor mutation burden (TMB) differentiates responders from nonresponders in some cases, it is a weak predictor in tumor types with low mutation rates. Thus, there is an unmet need to discover a new class of genetic aberrations that predict ICB responses in these tumor types. Here, we report analyses of pan-cancer whole genomes which revealed that intragenic rearrangement (IGR) burden is significantly associated with immune infiltration in breast, ovarian, esophageal, and endometrial cancers, particularly with increased M1 macrophage and CD8+ T-cell signatures. Multivariate regression against spatially counted tumor-infiltrating lymphocytes in breast, endometrial, and ovarian cancers suggested that IGR burden is a more influential covariate than other genetic aberrations in these cancers. In the MEDI4736 trial evaluating durvalumab in esophageal adenocarcinoma, IGR burden correlated with patient benefits. In the IMVigor210 trial evaluating atezolizumab in urothelial carcinoma, IGR burden increased with platinum exposure and predicted patient benefit among TMB-low, platinum-exposed tumors. Altogether, we have demonstrated that IGR burden correlates with T-cell inflammation and predicts ICB benefit in TMB-low, IGR-dominant tumors, and in platinum-exposed tumors.


Subject(s)
Carcinoma, Transitional Cell , Urinary Bladder Neoplasms , Female , Humans , Immune Checkpoint Inhibitors , Platinum , Biomarkers, Tumor/genetics , Mutation
2.
Oncogene ; 43(14): 1007-1018, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38361046

ABSTRACT

One-third of pediatric patients with osteosarcoma (OS) develop lung metastases (LM), which is the primary predictor of mortality. While current treatments of patients with localized bone disease have been successful in producing 5-year survival rates of 65-70%, patients with LM experience poor survival rates of only 19-30%. Unacceptably, this situation that has remained unchanged for 30 years. Thus, there is an urgent need to elucidate the mechanisms of metastatic spread in OS and to identify targetable molecular pathways that enable more effective treatments for patients with LM. We aimed to identify OS-specific gene alterations using RNA-sequencing of extremity and LM human tissues. Samples of extremity and LM tumors, including 4 matched sets, were obtained from patients with OS. Our data demonstrate aberrant regulation of the androgen receptor (AR) pathway in LM and predicts aldehyde dehydrogenase 1A1 (ALDH1A1) as a downstream target. Identification of AR pathway upregulation in human LM tissue samples may provide a target for novel therapeutics for patients with LM resistant to conventional chemotherapy.


Subject(s)
Bone Neoplasms , Lung Neoplasms , Osteosarcoma , Humans , Child , Aldehyde Dehydrogenase/metabolism , Receptors, Androgen/genetics , Lung Neoplasms/pathology , Osteosarcoma/pathology , Bone Neoplasms/pathology , RNA
3.
J STEM Outreach ; 5(2)2022.
Article in English | MEDLINE | ID: mdl-36910569

ABSTRACT

The University of Pittsburgh Medical Center Hillman Cancer Center Academy (Hillman Academy) has the primary goal of reaching high school students from underrepresented and disadvantaged backgrounds and guiding them through a cutting-edge research and professional development experience that positions them for success in STEM. With this focus, the Hillman Academy has provided nearly 300 authentic mentored research internship opportunities to 239 students from diverse backgrounds over the past 13 years most of whom matriculated into STEM majors in higher education. These efforts have helped shape a more diverse generation of future scientists and clinicians, who will enrich these fields with their unique perspectives and lived experiences. In this paper, we describe our program and the strategies that led to its growth into a National Institutes of Health Youth Enjoy Science-funded program including our unique multi-site structure, tiered mentoring platform, multifaceted recruitment approach, professional and academic development activities, and a special highlight of a set of projects with Deaf and Hard of Hearing students. We also share student survey data from the past six years that indicate satisfaction with the program, self-perceived gains in key areas of scientific development, awareness of careers in STEM, and an increased desire to pursue advanced degrees in STEM.

4.
J STEM Outreach ; 4(3)2021.
Article in English | MEDLINE | ID: mdl-35252781

ABSTRACT

In 2020, STEM training programs across the country were challenged to provide support to students during a nation-wide shutdown of research institutions in response to the COVID-19 pandemic. Five U.S. high school science internship programs funded by the Doris Duke Charitable Foundation, with a history of collaboration, developed innovative strategies for distance-learning (DL) opportunities during the pandemic. Forty under-represented high school and undergraduate students were paired with scientific mentors at one of the programs for a DL scientific internship. Summer training combined synchronous and asynchronous programming with research projects adapted for DL success. Ninety-five percent of students who participated were satisfied with the training experience, nearly identical to exit survey responses from 2019 when our programs were held in-person. More students were interested in pursuing a career in research at the end of the program and credited the DL experience with increasing interest in research careers. Some DL elements were ideal for underrepresented youth, including a more flexible schedule and elimination of cost and time for travel. While the lack of in-person instruction challenged our ability to create a strong student community, we found that preparation, communication, and flexibility were key elements to these successful DL programs. The increased emphasis on interpretation and analysis of data, rather than data collection, enhanced student learning. This manuscript highlights the changes made to our curricula, elements which were most successful, and recommends strategies for future distance-learning programming.

5.
Sci Rep ; 10(1): 8583, 2020 05 22.
Article in English | MEDLINE | ID: mdl-32444795

ABSTRACT

Evidence suggests Insulin-like growth factor 1 (IGF1) signaling is involved in the initiation and progression of a subset of breast cancers by inducing cell proliferation and survival. Although the signaling cascade following IGF1 receptor activation is well-studied, the key elements of the transcriptional response governing IGF1's actions are not well understood. Recent studies reveal that the majority of the genome is transcribed and that there are more long non-coding RNAs (lncRNAs) than protein coding genes, several of which are dysregulated in human cancer. However, studies on the regulation and mechanism of action of these lncRNAs are in their infancy. Here we show that IGF1 alters the expression levels of a subset of lncRNAs. SNHG7, a member of the small nucleolar host gene family, is a highly-expressed lncRNA that is consistently and significantly down-regulated by IGF1 signaling by a post-transcriptional mechanism through the MAPK pathway. SNHG7 regulates proliferation of breast cancer cell lines in a dose-dependent manner, and silencing SNHG7 expression causes cell cycle arrest in G0/G1. Intriguingly, SNHG7 alters the expression of many IGF1 signaling intermediates and IGF1-regulated genes suggesting a feedback mechanism to tightly regulate the IGF1 response. Finally, we show in clinical data that SNHG7 is overexpressed in tumors of a subset of breast cancer patients and that these patients have lower disease-free survival than patients without elevated SNHG7 expression. We propose that SNHG7 is a lncRNA oncogene that is controlled by growth factor signaling in a feedback mechanism to prevent hyperproliferation, and that this regulation can be lost in the development or progression of breast cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Cell Proliferation , Insulin-Like Growth Factor I/metabolism , RNA, Long Noncoding/genetics , Apoptosis , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Insulin-Like Growth Factor I/genetics , Prognosis , Survival Rate , Tumor Cells, Cultured
6.
Clin Cancer Res ; 24(20): 5165-5177, 2018 10 15.
Article in English | MEDLINE | ID: mdl-29941485

ABSTRACT

Purpose: Insulin-like growth factor 1 (IGF1) signaling regulates breast cancer initiation and progression and associated cancer phenotypes. We previously identified E-cadherin (CDH1) as a repressor of IGF1 signaling and in this study examined how loss of E-cadherin affects IGF1R signaling and response to anti-IGF1R/insulin receptor (InsR) therapies in breast cancer.Experimental Design: Breast cancer cell lines were used to assess how altered E-cadherin levels regulate IGF1R signaling and response to two anti-IGF1R/InsR therapies. In situ proximity ligation assay (PLA) was used to define interaction between IGF1R and E-cadherin. TCGA RNA-seq and RPPA data were used to compare IGF1R/InsR activation in estrogen receptor-positive (ER+) invasive lobular carcinoma (ILC) and invasive ductal carcinoma (IDC) tumors. ER+ ILC cell lines and xenograft tumor explant cultures were used to evaluate efficacy to IGF1R pathway inhibition in combination with endocrine therapy.Results: Diminished functional E-cadherin increased both activation of IGF1R signaling and efficacy to anti-IGF1R/InsR therapies. PLA demonstrated a direct endogenous interaction between IGF1R and E-cadherin at points of cell-cell contact. Increased expression of IGF1 ligand and levels of IGF1R/InsR phosphorylation were observed in E-cadherin-deficient ER+ ILC compared with IDC tumors. IGF1R pathway inhibitors were effective in inhibiting growth in ER+ ILC cell lines and synergized with endocrine therapy and similarly IGF1R/InsR inhibition reduced proliferation in ILC tumor explant culture.Conclusions: We provide evidence that loss of E-cadherin hyperactivates the IGF1R pathway and increases sensitivity to IGF1R/InsR targeted therapy, thus identifying the IGF1R pathway as a potential novel target in E-cadherin-deficient breast cancers. Clin Cancer Res; 24(20); 5165-77. ©2018 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Cadherins/metabolism , Drug Resistance, Neoplasm , Insulin-Like Growth Factor I/metabolism , Receptors, Somatomedin/metabolism , Signal Transduction/drug effects , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cadherins/genetics , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Synergism , Female , Gene Expression Profiling , Humans , Immunohistochemistry , Insulin-Like Growth Factor I/antagonists & inhibitors , Mice , RNA, Small Interfering/genetics , Receptor, IGF Type 1 , Receptors, Somatomedin/antagonists & inhibitors , Xenograft Model Antitumor Assays
7.
J Biol Chem ; 293(10): 3700-3709, 2018 03 09.
Article in English | MEDLINE | ID: mdl-29330302

ABSTRACT

Breast cancer development and progression are influenced by insulin-like growth factor receptor 1 (IGF1R) and insulin receptor (InsR) signaling, which drive cancer phenotypes such as cell growth, proliferation, and migration. IGF1R and InsR form IGF1R/InsR hybrid receptors (HybRs) consisting of one molecule of IGF1R and one molecule of InsR. The specific signaling and functions of HybR are largely unknown, as HybR is activated by both IGF1 and insulin, and no cellular system expresses HybR in the absence of holo-IGF1R or holo-InsR. Here we studied the role of HybR by constructing inducible chimeric receptors and compared HybR signaling with that of holo-IGF1R and holo-InsR. We cloned chemically inducible chimeric IGF1R and InsR constructs consisting of the extracellular domains of the p75 nerve growth factor receptor fused to the intracellular ß subunit of IGF1R or InsR and a dimerization domain. Dimerization with the drugs AP20187 or AP21967 allowed specific and independent activation of holo-IGF1R, holo-InsR, or HybR, resulting in activation of the PI3K pathway. Holo-IGF1R and HybR both promoted cell proliferation and glucose uptake, whereas holo-InsR only promoted glucose uptake, and only holo-IGF1R showed anti-apoptotic effects. We also found that the three receptors differentially regulated gene expression: holo-IGF1R and HybR up-regulated EGR3; holo-InsR specifically down-regulated JUN and BCL2L1; holo-InsR down-regulated but HybR up-regulated HK2; and HybR specifically up-regulated FHL2, ITGA6, and PCK2. Our findings suggest that, when expressed and activated in mammary epithelial cells, HybR acts in a manner similar to IGF1R and support further investigation of the role of HybR in breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Mammary Glands, Human/metabolism , Models, Molecular , Neoplasm Proteins/metabolism , Receptor, Insulin/metabolism , Receptors, Somatomedin/metabolism , Animals , Breast Neoplasms/pathology , Cell Line, Transformed , Cell Proliferation/drug effects , Female , Humans , Indicators and Reagents/pharmacology , Insulin/metabolism , Insulin-Like Growth Factor I/metabolism , MCF-7 Cells , Mammary Glands, Human/drug effects , Mammary Glands, Human/pathology , Mice , Neoplasm Proteins/agonists , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Protein Multimerization/drug effects , Receptor, Insulin/agonists , Receptor, Insulin/chemistry , Receptor, Insulin/genetics , Receptors, Somatomedin/agonists , Receptors, Somatomedin/chemistry , Receptors, Somatomedin/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Signal Transduction/drug effects , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , Tacrolimus/analogs & derivatives , Tacrolimus/pharmacology
8.
Curr Pathobiol Rep ; 5(1): 23-34, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28616363

ABSTRACT

PURPOSE OF REVIEW: The goal of this review was to compare and contrast the results and implications from several recent transcriptomic studies that analyzed the expression of lncRNAs in breast cancer. How many lncRNAs are dysregulated in breast cancer? Do dysregulated lncRNAs contribute to breast cancer etiology? Are lncRNAs viable biomarkers in breast cancer? RECENT FINDINGS: Transcriptomic profiling of breast cancer tissues, mostly from The Cancer Genome Atlas, identified thousands of long noncoding RNAs that are expressed and dysregulated in breast cancer. The expression of lncRNAs alone can divide patients into molecular subtypes. Subsequent functional studies demonstrated that several of these lncRNAs have important roles in breast cancer cell biology. SUMMARY: Thousands of lncRNAs are dysregulated in breast cancer that can be developed as biomarkers for prognostic or therapeutic purposes. The reviewed reports provide a roadmap to guide functional studies to discover lncRNAs with critical biological functions relating to breast cancer development and progression.

9.
J Pathol Inform ; 8: 2, 2017.
Article in English | MEDLINE | ID: mdl-28400991

ABSTRACT

The University of Pittsburgh's Department of Biomedical Informatics and Division of Pathology Informatics created a Science, Technology, Engineering, and Mathematics (STEM) pipeline in 2011 dedicated to providing cutting-edge informatics research and career preparatory experiences to a diverse group of highly motivated high-school students. In this third editorial installment describing the program, we provide a brief overview of the pipeline, report on achievements of the past scholars, and present results from self-reported assessments by the 2015 cohort of scholars. The pipeline continues to expand with the 2015 addition of the innovation internship, and the introduction of a program in 2016 aimed at offering first-time research experiences to undergraduates who are underrepresented in pathology and biomedical informatics. Achievements of program scholars include authorship of journal articles, symposium and summit presentations, and attendance at top 25 universities. All of our alumni matriculated into higher education and 90% remain in STEM majors. The 2015 high-school program had ten participating scholars who self-reported gains in confidence in their research abilities and understanding of what it means to be a scientist.

10.
Cell Rep ; 17(8): 2075-2086, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27851969

ABSTRACT

Cancer progression depends on both cell-intrinsic processes and interactions between different cell types. However, large-scale assessment of cell type composition and molecular profiles of individual cell types within tumors remains challenging. To address this, we developed epigenomic deconvolution (EDec), an in silico method that infers cell type composition of complex tissues as well as DNA methylation and gene transcription profiles of constituent cell types. By applying EDec to The Cancer Genome Atlas (TCGA) breast tumors, we detect changes in immune cell infiltration related to patient prognosis, and a striking change in stromal fibroblast-to-adipocyte ratio across breast cancer subtypes. Furthermore, we show that a less adipose stroma tends to display lower levels of mitochondrial activity and to be associated with cancerous cells with higher levels of oxidative metabolism. These findings highlight the role of stromal composition in the metabolic coupling between distinct cell types within tumors.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Epigenomics , Adipose Tissue/pathology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Line, Tumor , Computer Simulation , DNA Methylation/genetics , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Humans , Oxidation-Reduction , Phenotype , Reproducibility of Results , Sequence Analysis, DNA , Stromal Cells/pathology , Tumor Microenvironment/genetics
11.
Article in English | MEDLINE | ID: mdl-25964777

ABSTRACT

Insulin-like growth factor (IGF) signaling is fundamental for growth and survival. A large body of evidence (laboratory, epidemiological, and clinical) implicates the exploitation of this pathway in cancer. Up to 50% of breast tumors express the activated form of the type 1 insulin-like growth factor receptor (IGF1R). Breast cancers are categorized into subtypes based upon hormone and ERRB2 receptor expression and/or gene expression profiling. Even though IGF1R influences tumorigenic phenotypes and drug resistance across all breast cancer subtypes, it has specific expression and function in each. In some subtypes, IGF1R levels correlate with a favorable prognosis, while in others it is associated with recurrence and poor prognosis, suggesting different actions based upon cellular and molecular contexts. In this review, we examine IGF1R expression and function as it relates to breast cancer subtype and therapy-acquired resistance. Additionally, we discuss the role of IGF1R in stem cell maintenance and lineage differentiation and how these cell fate influences may alter the differentiation potential and cellular composition of breast tumors.

12.
Proc Natl Acad Sci U S A ; 110(3): 978-83, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23277542

ABSTRACT

The oncogenic transcription factor c-Myc causes transformation and tumorigenesis, but it can also induce apoptotic cell death. Although tumor suppressors are necessary for c-Myc to induce apoptosis, the pathways and mechanisms are unclear. To further understand how c-Myc switches from an oncogenic protein to an apoptotic protein, we examined the mechanism of p53-independent c-Myc-induced apoptosis. We show that the tumor suppressor protein ARF mediates this switch by inhibiting ubiquitylation of the c-Myc transcriptional domain (TD). Whereas TD ubiquitylation is critical for c-Myc canonical transcriptional activity and transformation, inhibition of ubiquitylation leads to the induction of the noncanonical c-Myc target gene, Egr1, which is essential for efficient c-Myc-induced p53-independent apoptosis. ARF inhibits the interaction of c-Myc with the E3 ubiquitin ligase Skp2. Overexpression of Skp2, which occurs in many human tumors, inhibits the recruitment of ARF to the Egr1 promoter, leading to inhibition of c-Myc-induced apoptosis. Therapeutic strategies could be developed to activate this intrinsic apoptotic activity of c-Myc to inhibit tumorigenesis.


Subject(s)
Apoptosis/physiology , Proto-Oncogene Proteins c-myc/chemistry , Proto-Oncogene Proteins c-myc/metabolism , Amino Acid Substitution , Animals , Apoptosis/genetics , Binding, Competitive , Cell Transformation, Neoplastic , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Early Growth Response Protein 1/genetics , Gene Knockout Techniques , Genes, myc , HeLa Cells , Humans , Mice , Mutagenesis, Site-Directed , Protein Structure, Tertiary , Proto-Oncogene Proteins c-myc/genetics , S-Phase Kinase-Associated Proteins/metabolism , Transcription, Genetic , Ubiquitination
13.
Crit Rev Oncog ; 17(2): 161-73, 2012.
Article in English | MEDLINE | ID: mdl-22471706

ABSTRACT

Overwhelming evidence implicates insulin-like growth factor (IGF) signaling in the growth and survival of many types of human cancer cells. Numerous inhibitors of the IGF1 receptor (IGF1R) have been developed, and they displayed remarkable antineoplastic activity in preclinical models and promising success in early phase clinical trials. However, while responses have been observed in numerous cancer types, they have occurred in a minority of patients, and serious toxicities have been observed. Identifying patients likely to benefit from anti-IGF1R therapy requires further characterizing the role of IGF1 signaling in various stages of tumorigenesis in order to identify critical downstream factors that may be used as predictors of response, or to serve as novel therapeutic targets. Recent microarray analyses have begun to unravel expression "signatures" specific for IGF1 that correlate with poor breast cancer prognosis and with response to anti-IGFIR inhibitors. In this review we briefly discuss the history of the IGF1 family in neoplasia, how it is targeted, results from clinical trials, and the quest for biomarkers that will predict response to IGF1R-targeted therapy.


Subject(s)
Biomarkers/metabolism , Gene Expression Profiling , Receptors, Somatomedin/drug effects , Humans , Insulin-Like Growth Factor I/metabolism , Signal Transduction , Transcription, Genetic
15.
Proc Natl Acad Sci U S A ; 108(2): 632-7, 2011 Jan 11.
Article in English | MEDLINE | ID: mdl-21187408

ABSTRACT

c-Myc is frequently deregulated in human cancers. Although deregulated c-Myc leads to tumor growth, it also triggers apoptosis in partnership with tumor suppressors such as ARF and p53. Apoptosis induced by c-Myc is a critical fail-safe mechanism for the cell to protect against unrestrained proliferation. Despite the plethora of information on c-Myc, the molecular mechanism of how c-Myc induces both transformation and apoptosis is unclear. Oncogenic c-Myc can indirectly induce the expression of the tumor suppressor ARF, which leads to apoptosis through the stabilization of p53, but both c-Myc and ARF have apoptotic activities that are independent of p53. In cells without p53, ARF directly binds to c-Myc protein and inhibits c-Myc-induced hyperproliferation and transformation with a concomitant inhibition of canonical c-Myc target gene induction. However, ARF is an essential cofactor for p53-independent c-Myc-induced apoptosis. Here we show that ARF is necessary for c-Myc to drive transcription of a unique noncanonical target gene, Egr1. In contrast, c-Myc induces another family member, Egr2, through a canonical mechanism that is inhibited by ARF. We further demonstrate that Egr1 is essential for p53-independent c-Myc-induced apoptosis, but not ARF-independent c-Myc-induced apoptosis. Therefore, ARF binding switches the inherent activity of c-Myc from a proliferative to apoptotic protein without p53 through a unique noncanonical transcriptional mechanism. These findings also provide evidence that cofactors can differentially regulate specific transcriptional programs of c-Myc leading to different biological outcomes.


Subject(s)
ADP-Ribosylation Factors/metabolism , Apoptosis , Early Growth Response Protein 1/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Animals , Binding Sites , COS Cells , Chlorocebus aethiops , Chromatin/chemistry , Fibroblasts/metabolism , Mice , Mice, Transgenic , RNA Interference , Rats
16.
Mol Biol Evol ; 23(3): 587-97, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16291999

ABSTRACT

Morphological data supports monotremes as the sister group of Theria (extant marsupials + eutherians), but phylogenetic analyses of 12 mitochondrial protein-coding genes have strongly supported the grouping of monotremes with marsupials: the Marsupionta hypothesis. Various nuclear genes tend to support Theria, but a comprehensive study of long concatenated sequences and broad taxon sampling is lacking. We therefore determined sequences from six nuclear genes and obtained additional sequences from the databases to create two large and independent nuclear data sets. One (data set I) emphasized taxon sampling and comprised five genes, with a concatenated length of 2,793 bp, from 21 species (two monotremes, six marsupials, nine placentals, and four outgroups). The other (data set II) emphasized gene sampling and comprised eight genes and three proteins, with a concatenated length of 10,773 bp or 3,669 amino acids, from five taxa (a monotreme, a marsupial, a rodent, human, and chicken). Both data sets were analyzed by parsimony, minimum evolution, maximum likelihood, and Bayesian methods using various models and data partitions. Data set I gave bootstrap support values for Theria between 55% and 100%, while support for Marsupionta was at most 12.3%. Taking base compositional bias into account generally increased the support for Theria. Data set II exclusively supported Theria, with the highest possible values and significantly rejected Marsupionta. Independent phylogenetic evidence in support of Theria was obtained from two single amino acid deletions and one insertion, while no supporting insertions and deletions were found for Marsupionta. On the basis of our data sets, the time of divergence between Monotremata and Theria was estimated at 231-217 MYA and between Marsupialia and Eutheria at 193-186 MYA. The morphological evidence for a basal position of Monotremata, well separated from Theria, is thus fully supported by the available molecular data from nuclear genes.


Subject(s)
Cell Nucleus/genetics , Classification , Phylogeny , Platypus , Amino Acid Sequence , Animals , Biological Evolution , Codon , Data Interpretation, Statistical , Humans , Marsupialia/classification , Marsupialia/genetics , Molecular Sequence Data , Platypus/classification , Platypus/genetics , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL
...