Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(9)2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38731959

ABSTRACT

Cerebral cavernous malformations (CCMs) are a neurological disorder characterized by enlarged intracranial capillaries in the brain, increasing the susceptibility to hemorrhagic strokes, a major cause of death and disability worldwide. The limited treatment options for CCMs underscore the importance of prognostic biomarkers to predict the likelihood of hemorrhagic events, aiding in treatment decisions and identifying potential pharmacological targets. This study aimed to identify blood biomarkers capable of diagnosing and predicting the risk of hemorrhage in CCM1 patients, establishing an initial set of circulating biomarker signatures. By analyzing proteomic profiles from both human and mouse CCM models and conducting pathway enrichment analyses, we compared groups to identify potential blood biomarkers with statistical significance. Specific candidate biomarkers primarily associated with metabolism and blood clotting pathways were identified. These biomarkers show promise as prognostic indicators for CCM1 deficiency and the risk of hemorrhagic stroke, strongly correlating with the likelihood of hemorrhagic cerebral cavernous malformations (CCMs). This lays the groundwork for further investigation into blood biomarkers to assess the risk of hemorrhagic CCMs.


Subject(s)
Biomarkers , Hemangioma, Cavernous, Central Nervous System , Hemangioma, Cavernous, Central Nervous System/blood , Hemangioma, Cavernous, Central Nervous System/diagnosis , Humans , Animals , Mice , Prognosis , Biomarkers/blood , Proteomics/methods , Cerebral Hemorrhage/blood , Cerebral Hemorrhage/diagnosis , KRIT1 Protein/blood , Disease Models, Animal , Female , Male
2.
Diagnostics (Basel) ; 13(6)2023 Mar 07.
Article in English | MEDLINE | ID: mdl-36980321

ABSTRACT

Liver cancer, comprising hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA), is a leading cause of cancer-related deaths worldwide. The liver is a primary metabolic organ for progesterone (PRG) and PRG exerts its effects through classic nuclear PRG receptors (nPRs) and non-classic membrane PRG receptors (mPRs) or a combination of both. Previous studies have shown that the CCM signaling complex (CSC) couples both nPRs and mPRs to form the CmPn (CSC-mPR-PRG-nPR) signaling network, which is involved in multiple cellular signaling pathways, including tumorigenesis of various cancers. Despite advances in treatment, 5-year survival rates for liver cancer patients remain low, largely due to the chemoresistant nature of HCCs. The lack of sensitive and specific biomarkers for liver cancer diagnosis and prognosis emphasizes the need for identifying new potential biomarkers. We propose the potential use of CmPn members' expression data as prognostic biomarkers or biomarker signatures for the major types of hepatic cancer, including HCCs and CCAs, as well as rare subtypes such as undifferentiated pleomorphic sarcoma (UPS) and hepatic angiosarcoma (HAS). In this study, we investigated the CmPn network through RNAseq data and immunofluorescence techniques to measure alterations to key cancer pathways during liver tumorigenesis. Our findings reveal significant differential expression of multiple CmPn members, including CCM1, PAQR7, PGRMC1, and nPRs, in both HCCs and CCAs, highlighting the crucial roles of mPRs, nPRs, and CSC signaling during liver tumorigenesis. These key members of the CmPn network may serve as potential biomarkers for the diagnosis and prognosis of liver cancer subtypes, including rare subtypes.

3.
Int J Mol Sci ; 23(17)2022 Aug 26.
Article in English | MEDLINE | ID: mdl-36077089

ABSTRACT

Cerebral cavernous malformations (CCMs) are characterized by abnormally dilated intracranial microvascular sinusoids that result in increased susceptibility to hemorrhagic stroke. It has been demonstrated that three CCM proteins (CCM1, CCM2, and CCM3) form the CCM signaling complex (CSC) to mediate angiogenic signaling. Disruption of the CSC will result in hemorrhagic CCMs, a consequence of compromised blood-brain barrier (BBB) integrity. Due to their characteristically incomplete penetrance, the majority of CCM mutation carriers (presumed CCM patients) are largely asymptomatic, but when symptoms occur, the disease has typically reached a clinical stage of focal hemorrhage with irreversible brain damage. We recently reported that the CSC couples both classic (nuclear; nPRs) and nonclassic (membrane; mPRs) progesterone (PRG)-receptors-mediated signaling within the CSC-mPRs-PRG (CmP) signaling network in nPR(-) breast cancer cells. In this report, we demonstrate that depletion of any of the three CCM genes or treatment with mPR-specific PRG actions (PRG/mifepristone) results in the disruption of the CmP signaling network, leading to increased permeability in the nPR(-) endothelial cells (ECs) monolayer in vitro. Finally, utilizing our in vivo hemizygous Ccm mutant mice models, we demonstrate that depletion of any of the three CCM genes, in combination with mPR-specific PRG actions, is also capable of leading to defective homeostasis of PRG in vivo and subsequent BBB disruption, allowing us to identify a specific panel of etiological blood biomarkers associated with BBB disruption. To our knowledge, this is the first report detailing the etiology to predict the occurrence of a disrupted BBB, an indication of early hemorrhagic events.


Subject(s)
Endothelial Cells , Hemangioma, Cavernous, Central Nervous System , Animals , Blood-Brain Barrier/metabolism , Cytidine Monophosphate/metabolism , Endothelial Cells/metabolism , Hemangioma, Cavernous, Central Nervous System/genetics , Mice , Microtubule-Associated Proteins/metabolism , Signal Transduction
4.
Cell Commun Signal ; 20(1): 120, 2022 08 15.
Article in English | MEDLINE | ID: mdl-35971177

ABSTRACT

BACKGROUND: Breast cancer, the most diagnosed cancer, remains the second leading cause of cancer death in the United States, and excessive Progesterone (PRG) or Mifepristone (MIF) exposure may be at an increased risk for developing breast cancer. PRG exerts its cellular responses through signaling cascades involving classic, non-classic, or combined responses by binding to either classic nuclear PRG receptors (nPRs) or non-classic membrane PRG receptors (mPRs). Currently, the intricate balance and switch mechanisms between these two signaling cascades remain elusive. Three genes, CCM1-3, form the CCM signaling complex (CSC) which mediates multiple signaling cascades. METHODS: Utilizing molecular, cellular, Omics, and systems biology approaches, we analyzed the relationship among the CSC, PRG, and nPRs/mPRs during breast cancer tumorigenesis. RESULTS: We discovered that the CSC plays an essential role in coupling both classic and non-classic PRG signaling pathways by mediating crosstalk between them, forming the CmPn (CSC-mPRs-PRG-nPRs) signaling network. We found that mPR-specific PRG actions (PRG + MIF) play an essential role in this CmPn network during breast cancer tumorigenesis. Additionally, we have identified 4 categories of candidate biomarkers (9 intrinsic, 2 PRG-inducible, 1 PRG-repressive, 1 mPR-specific PRG-repressive, and 2 mPR-responsive) for Luminal-A breast cancers during tumorigenesis and have confirmed the prognostic application of RPL13 and RPL38 as intrinsic biomarkers using a dual validation method. CONCLUSIONS: We have discovered that the CSC plays an essential role in the CmPn signaling network for Luminal-A breast cancers with identification of two intrinsic biomarkers. Video Abstract.


Subject(s)
Breast Neoplasms , Receptors, Progesterone , Carcinogenesis , Female , Humans , Neoplasm Proteins/metabolism , Progesterone/metabolism , Receptors, Progesterone/metabolism , Ribosomal Proteins/metabolism , Signal Transduction
5.
Cancer Biomark ; 34(4): 607-636, 2022.
Article in English | MEDLINE | ID: mdl-35431232

ABSTRACT

Breast cancer is the most diagnosed cancer worldwide and remains the second leading cause of cancer death. While breast cancer mortality has steadily declined over the past decades through medical advances, an alarming disparity in breast cancer mortality has emerged between African American women (AAW) and Caucasian American women (CAW). New evidence suggests more aggressive behavior of triple-negative breast cancer (TNBC) in AAW may contribute to racial differences in tumor biology and mortality. Progesterone (PRG) can exert its cellular effects through either its classic, non-classic, or combined responses through binding to either classic nuclear PRG receptors (nPRs) or non-classic membrane PRG receptors (mPRs), warranting both pathways equally important in PRG-mediated signaling. In our previous report, we demonstrated that the CCM signaling complex (CSC) consisting of CCM1, CCM2, and CCM3 can couple both nPRs and mPRs signaling cascades to form a CSC-mPRs-PRG-nPRs (CmPn) signaling network in nPR positive(+) breast cancer cells. In this report, we furthered our research by establishing the CSC-mPRs-PRG (CmP) signaling network in nPR(-) breast cancer cells, demonstrating that a common core mechanism exists, regardless of nPR(+⁣/⁣-) status. This is the first report stating that inducible expression patterns exist between CCMs and major mPRs in TNBC cells. Furthermore, we firstly show mPRs in TNBC cells are localized in the nucleus and participate in nucleocytoplasmic shuttling in a coordinately synchronized fashion with CCMs under steroid actions, following the same cellular distribution as other well-defined steroid hormone receptors. Finally, for the first time, we deconvoluted the CmP signalosome by using systems biology and TNBC clinical data, which helped us understand key factors within the CmP network and identify 6 specific biomarkers with potential clinical applications associated with AAW-TNBC tumorigenesis. These novel biomarkers could have immediate clinical implications to dramatically improve health disparities among AAW-TNBCs.


Subject(s)
Triple Negative Breast Neoplasms , Black or African American , Biomarkers, Tumor/metabolism , Female , Humans , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , White People
6.
Genet Test Mol Biomarkers ; 26(4): 198-219, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35481969

ABSTRACT

Objective: Triple-negative breast cancer (TNBC) constitutes ∼15% of all diagnosed invasive breast cancer cases with limited options for treatment since immunotherapies that target ER, PR, and HER2 receptors are ineffective. Progesterone (PRG) can induce its effects through either classic, nonclassic, or combined responses by binding to classic nuclear PRG receptors (nPRs) or nonclassic membrane PRG receptors (mPRs). Under PRG-induced actions, we previously demonstrated that the CCM signaling complex (CSC) can couple both nPRs and mPRs into a CmPn signaling network, which plays an important role during nPR(+) breast cancer tumorigenesis. We recently defined the novel CmP signaling network in African American women (AAW)-derived TNBC cells, which overlapped with our previously defined CmPn network in nPR(+) breast cancer cells. Methods: Under mPR-specific steroid actions, we measured alterations to key tumorigenic pathways in Caucasian American women (CAW)- derived TNBC cells, with RNAseq/proteomic and systems biology approaches. Exemption from ethics approval from IRB: This study only utilized cultured NBC cell lines with publicly available TNBC clinical data sets. Results: Our results demonstrated that TNBCs in CAW share similar altered signaling pathways, as TNBCs in AAW, under mPR-specific steroid actions, demonstrating the overall aggressive nature of TNBCs, regardless of racial differences. Furthermore, in this report, we have deconvoluted the CmP signalosome, using systems biology approaches and CAW-TNBC clinical data, to identify 21 new CAW-TNBC-specific prognostic biomarkers that reinforce the definitive role of CSC and mPR signaling during CAW-TNBC tumorigenesis. Conclusion: This new set of potential prognostic biomarkers may revolutionize molecular mechanisms and currently known concepts of tumorigenesis in CAW-TNBCs, leading to hopeful new therapeutic strategies.


Subject(s)
Triple Negative Breast Neoplasms , Biomarkers , Carcinogenesis , Female , Humans , Prognosis , Proteomics , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism
7.
Article in English | MEDLINE | ID: mdl-35477890

ABSTRACT

Somatic gain-of-function (GOF) mutations in phosphatidylinositol-4, 5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), the catalytic subunit of phosphoinositide 3-kinase (PI3K), have been recently discovered in cerebral cavernous malformations (CCMs), raising the possibility that the activation of PI3K pathways is a possible universal regulator of vascular morphogenesis. However, there have been contradicting data presented among various groups and studies. To enhance the current understanding of vascular anomalies, it is essential to explore this possible relationship between altered PI3K signalling pathways and its influence on the pathogenesis of CCMs. GOF PIK3CA-mutants have been linked to overgrowth syndromes, allowing this group of disorders, resulting from somatic activating mutations in PIK3CA, to be collectively named as PIK3CA-related overgrowth spectrum disorders. This paper reviews and attempts to conceptualise the relationships and differences among clinical presentations, genotypic and phenotypic correlations and possible coexistence of PIK3CA and CCM mutations/phenotypes in CCM lesions. Finally, we present a model reflecting our hypothetical understanding of CCM pathogenesis based on a systematic review and conceptualisation of data obtained from other studies.

8.
Biochem Biophys Rep ; 29: 101218, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35128084

ABSTRACT

PURPOSE: The objective of this study is to validate the existence of dual cores within the typical phosphotyrosine binding (PTB) domain and to identify potentially damaging and pathogenic nonsynonymous coding single nuclear polymorphisms (nsSNPs) in the canonical PTB domain of the CCM2 gene that causes cerebral cavernous malformations (CCMs). METHODS: The nsSNPs within the coding sequence for PTB domain of human CCM2 gene, retrieved from exclusive database searches, were analyzed for their functional and structural impact using a series of bioinformatic tools. The effects of mutations on the tertiary structure of the PTB domain in human CCM2 protein were predicted to examine the effect of nsSNPs on the tertiary structure of PTB Cores. RESULTS: Our mutation analysis, through alignment of protein structures between wildtype CCM2 and mutant, predicted that the structural impacts of pathogenic nsSNPs is biophysically limited to only the spatially adjacent substituted amino acid site with minimal structural influence on the adjacent core of the PTB domain, suggesting both cores are independently functional and essential for proper CCM2 PTB function. CONCLUSION: Utilizing a combination of protein conservation and structure-based analysis, we analyzed the structural effects of inherited pathogenic mutations within the CCM2 PTB domain. Our results predicted that the pathogenic amino acid substitutions lead to only subtle changes locally, confined to the surrounding tertiary structure of the PTB core within which it resides, while no structural disturbance to the neighboring PTB core was observed, reaffirming the presence of independently functional dual cores in the CCM2 typical PTB domain.

9.
Biochim Biophys Acta Gen Subj ; 1865(11): 129977, 2021 11.
Article in English | MEDLINE | ID: mdl-34391832

ABSTRACT

BACKGROUND: Phosphotyrosine Binding (PTB) Domains, usually found on scaffold proteins, are pervasive in many cellular signaling pathways. These domains are the second-largest family of phosphotyrosine recognition domains and since their initial discovery, dozens of PTB domains have been structurally determined. SCOPE OF REVIEW: Due to its signature sequence flexibility, PTB domains can bind to a large variety of ligands including phospholipids. PTB peptide binding is divided into classical binding (canonical NPXY motifs) and non-classical binding (all other motifs). The first atypical PTB domain was discovered in cerebral cavernous malformation 2 (CCM2) protein, while only one third in size of the typical PTB domain, it remains functionally equivalent. MAJOR CONCLUSIONS: PTB domains are involved in numerous signaling processes including embryogenesis, neurogenesis, and angiogenesis, while dysfunction is linked to major disorders including diabetes, hypercholesterolemia, Alzheimer's disease, and strokes. PTB domains may also be essential in infectious processes, currently responsible for the global pandemic in which viral cellular entry is suspected to be mediated through PTB and NPXY interactions. GENERAL SIGNIFICANCE: We summarize the structural and functional updates in the PTB domain over the last 20 years in hopes of resurging interest and further analyzing the importance of this versatile domain.


Subject(s)
Alzheimer Disease/metabolism , Diabetes Mellitus/metabolism , Hypercholesterolemia/metabolism , Phosphotyrosine/metabolism , Stroke/metabolism , Binding Sites , Humans , Phosphotyrosine/chemistry
10.
Vessel Plus ; 52021.
Article in English | MEDLINE | ID: mdl-35098046

ABSTRACT

Cerebral cavernous malformations (CCMs), one of the most common vascular malformations, are characterized by abnormally dilated intracranial microvascular capillaries resulting in increased susceptibility to hemorrhagic stroke. As an autosomal dominant disorder with incomplete penetrance, the majority of CCMs gene mutation carriers are largely asymptomatic but when symptoms occur, the disease has typically reached the stage of focal hemorrhage with irreversible brain damage, while the molecular "trigger" initiating the occurrence of CCM pathology remain elusive. Currently, the invasive neurosurgery removal of CCM lesions is the only option for the treatment, despite the recurrence of the worse symptoms frequently occurring after surgery. Therefore, there is a grave need for identification of molecular targets for therapeutic treatment and biomarkers as risk predictors for hemorrhagic stroke prevention. Based on reported various perturbed angiogenic signaling cascades mediated by the CCM signaling complex (CSC), there have been many proposed candidate drugs, targeting potentially angiogenic-relevant signaling pathways dysregulated by loss of function of one of the CCM proteins, which might not be enough to correct the pathological phenotype, hemorrhagic CCMs. In this review, we describe a new paradigm for the mechanism of hemorrhagic CCM lesions, and propose a new concept for the assurance of the CSC-stability to prevent the devastating outcome of hemorrhagic CCMs.

11.
Chin Neurosurg J ; 6: 4, 2020.
Article in English | MEDLINE | ID: mdl-32922933

ABSTRACT

BACKGROUND: Cerebral cavernous malformations (CCMs), a major neurosurgical condition, characterized by abnormally dilated intracranial capillaries, result in increased susceptibility to stroke. KRIT1 (CCM1), MGC4607 (CCM2), and PDCD10 (CCM3) have been identified as causes of CCMs in which at least one of them is disrupted in most familial cases. Our goal is to identify potential biomarkers and genetic modifiers of CCMs, using a global comparative omics approach across several in vitro studies and multiple in vivo animal models. We hypothesize that through analysis of the CSC utilizing various omics, we can identify potential biomarkers and genetic modifiers, by systemically evaluating effectors and binding partners of the CSC as well as second layer interactors. METHODS: We utilize a comparative omics approach analyzing multiple CCMs deficient animal models across nine independent studies at the genomic, transcriptomic, and proteomic levels to dissect alterations in various signaling cascades. RESULTS: Our analysis revealed a large set of genes that were validated across multiple independent studies, suggesting an important role for these identified genes in CCM pathogenesis. CONCLUSION: This is currently one of the largest comparative omics analysis of CCM deficiencies across multiple models, allowing us to investigate global alterations among multiple signaling cascades involved in both angiogenic and non-angiogenic events and to also identify potential biomarker candidates of CCMs, which can be used for new therapeutic strategies.

12.
Methods Mol Biol ; 2152: 225-251, 2020.
Article in English | MEDLINE | ID: mdl-32524556

ABSTRACT

The use of vertebrate models allows researchers to investigate mechanisms of CCM pathogenesis in vivo, to investigate discrepancies between observations seen in the lab with in vitro experiments and how they translate into animal models; these in vivo models are more relevant in terms of CCM pathogenesis seen in humans than the in vitro counterparts. The use of CCM-deficient Zebrafish model offers advantages given their optical clarity during embryogenesis, short generation time, and high fecundity. When looking at the in vivo mouse model, gene conservation among CCM1, CCM2, and CCM3 is much higher among mammals (>92%), offering higher relevance in terms of similarities between what is seen in a mouse compared to human CCM pathogenesis. With both models, deficiencies in CCM1, CCM2, and CCM3 demonstrate perturbed cardiovascular development and underlying mechanisms of CCM pathogenesis at multiple stages seen in humans. The optimized methods described in this chapter allow researchers to benefit from both in vivo models, investigating impacts of deficiencies in CCM gene expression and its effect on angiogenesis and other signaling cascades, offering a much wider view of the molecular and cellular mechanisms in CCM progression.


Subject(s)
Disease Models, Animal , Disease Susceptibility , Hemangioma, Cavernous, Central Nervous System/etiology , Hemangioma, Cavernous, Central Nervous System/metabolism , Alleles , Animals , Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Fluorescent Antibody Technique , Genomics/methods , Hemangioma, Cavernous, Central Nervous System/pathology , Immunophenotyping , In Situ Hybridization , Mice , Mice, Knockout , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mutation , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Phenotype , Proteomics/methods , Vertebrates , Zebrafish
13.
Methods Mol Biol ; 2152: 311-324, 2020.
Article in English | MEDLINE | ID: mdl-32524562

ABSTRACT

Molecular techniques allow for the rapid discovery of CCM-associated protein targets, crucial to understanding CCM pathogenesis. Here, we describe optimized protein extraction methods that allow for extraction from whole cell, and/or cellular sub-compartments, including nuclear, mitochondria, cytoplasmic, and membrane-bound proteins, from lysates. This allows for the analysis of in vitro co-immunoprecipitation (Co-IP), label-free measurement of protein-protein interactions, multiplex protein-lipid binding assays, and western blots. Together, all these methods allow for a global analysis of the molecular mechanisms underlying CCM pathogenesis.


Subject(s)
Hemangioma, Cavernous, Central Nervous System/etiology , Hemangioma, Cavernous, Central Nervous System/metabolism , Proteome , Proteomics , Carrier Proteins , Chromatography, Affinity , Humans , Immunoprecipitation , Lipids , Protein Binding , Protein Interaction Mapping , Proteomics/instrumentation , Proteomics/methods
14.
Methods Mol Biol ; 2152: 303-310, 2020.
Article in English | MEDLINE | ID: mdl-32524561

ABSTRACT

Cellular techniques allow researchers to discover underlying mechanisms of pathogenesis of CCMs in vitro before carrying over into in vivo models; optimization of these techniques facilitates the rapid discovery of CCM-associated gene and protein targets. Here, we describe optimized cell culture applications which are essential for successful molecular techniques and will offer researchers effective methods for plasmid transfections, facilitating mammalian cell expression, subcellular localization, and fluorescence microscopy. RNA interference (RNAi) treatment of cells allows for various in vitro cellular assays as well as confocal microscopy experiments. Together, all these methods allow for an in-depth analysis of the cellular mechanisms underlying CCM pathogenesis to be explored and further dissected.


Subject(s)
Disease Susceptibility , Hemangioma, Cavernous, Central Nervous System/etiology , Hemangioma, Cavernous, Central Nervous System/pathology , Microscopy , Biomarkers , Cell Line , Endothelial Cells/metabolism , Gene Expression , Humans , Microscopy/instrumentation , Microscopy/methods , Microscopy, Confocal , Microscopy, Fluorescence , Plasmids/genetics , Protein Transport , RNA, Small Interfering/genetics , Transfection
15.
Methods Mol Biol ; 2152: 325-344, 2020.
Article in English | MEDLINE | ID: mdl-32524563

ABSTRACT

Omics research has garnered popularity recently to integrate in-depth analysis of alterations at the molecular level to elucidate observable phenotypes resulting from knockdown/knockout models. Genomics, performed through RNA-seq, allows the user to evaluate alterations at the transcription level, oftentimes more sensitive than other types of analysis, especially when attempting to understand lack of observation of an expected phenotype. Proteomics facilitates an understanding of mechanisms being altered at the translational level allowing for an understanding of multiple layers of regulation occurring, elucidating discrepancies between what is seen at the RNA level compared to what is translated to a functional protein. Here we describe the methods currently being used to evaluate CCM-deficient strains in human brain microvascular endothelial cells (HBMVEC), zebrafish embryos as well as in vivo mouse model to evaluate impacts on various signaling cascades resulting from deficiencies in KRIT1 (CCM1), MGC4607 (CCM2), and PDCD10 (CCM3). The integration of data from genomics and proteomics analysis allows for the composition of interactomes, elucidating systems wide impacts resulting from disruption of the CCM signaling complex (CSC).


Subject(s)
Genomics , Hemangioma, Cavernous, Central Nervous System/etiology , Hemangioma, Cavernous, Central Nervous System/metabolism , Microtubule-Associated Proteins/metabolism , Proteomics , Signal Transduction , Animals , Chromatin Immunoprecipitation Sequencing , Chromatography, Liquid , Computational Biology/instrumentation , Computational Biology/methods , Disease Models, Animal , Genomics/instrumentation , Genomics/methods , Humans , Mice , Microtubule-Associated Proteins/genetics , Protein Binding , Protein Interaction Mapping , Proteomics/instrumentation , Proteomics/methods , Tandem Mass Spectrometry , Zebrafish
16.
Oncol Rep ; 43(6): 1945-1963, 2020 06.
Article in English | MEDLINE | ID: mdl-32186778

ABSTRACT

Cerebral cavernous malformations (CCMs) are microvascular anomalies in the brain that result in increased susceptibility to stroke. Three genes have been identified as causes of CCMs: cerebral cavernous malformations 1 [(CCM1) also termed Krev interaction trapped 1 (KRIT1)], cerebral cavernous malformation 2 [(CCM2) also termed MGC4607] and cerebral cavernous malformation 3 [(CCM3) also termed programmed cell death 10 (PDCD10)]. It has been demonstrated that both CCM1 and CCM3 bind to CCM2 to form a CCM signaling complex (CSC) with which to modulate multiple signaling cascades. CCM proteins have been reported to play major roles in microvascular angiogenesis in human and animal models. However, CCM proteins are ubiquitously expressed in all major tissues, suggesting an unseen broader role of the CSC in biogenesis. Recent evidence suggests the possible involvement of the CSC complex during tumorigenesis; however, studies concerning this aspect are limited. This is the first report to systematically investigate the expression patterns of CCM proteins in major human tumors using real­time quantitative PCR, RNA­fluorescence in situ hybridization, immunohistochemistry and multicolor immunofluorescence imaging. Our data demonstrated that differential expression patterns of the CSC complex are correlated with certain types and grades of major human cancers, indicating the potential application of CCM genes as molecular biomarkers for clinical oncology. Our data strongly suggest that more efforts are needed to elucidate the role of the CSC complex in tumorigenesis, which may have enormous clinical potential for cancer diagnostic, prognostic and therapeutic applications.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Carrier Proteins/genetics , KRIT1 Protein/genetics , Membrane Proteins/genetics , Neoplasms/genetics , Proto-Oncogene Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carrier Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , KRIT1 Protein/metabolism , Membrane Proteins/metabolism , Neoplasm Grading , Neoplasms/metabolism , Neoplasms/pathology , Proto-Oncogene Proteins/metabolism
17.
Biochem Biophys Res Commun ; 524(3): 595-607, 2020 04 09.
Article in English | MEDLINE | ID: mdl-32029278

ABSTRACT

Current understanding of phosphotyrosine binding (PTB) domain is limited. Recently, we revealed a novel atypical phosphotyrosine binding (aPTB) domain in CCM2, making it a dual PTB domain-containing protein. Since aPTB domain is only 1/3 of the size of typical PTB domain, we explored the possibility to decrease the size of PTB domain and demonstrate that the typical PTB domain can be divided into two similarly structural and functional cores that can independently bind to NPXY motif. Further, we reduced each PTB core into a minimum core motif (mCore) which is the functional unit of PTB domains and structurally similar to the novel aPTB domain. Based on structural data, we developed several cis- and trans-inhibitors for NPXY binding scheme, with potential applications for therapeutic strategies in human health conditions.


Subject(s)
Phosphotyrosine/chemistry , Phosphotyrosine/metabolism , Amino Acid Motifs , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Kinetics , Protein Binding , Protein Domains , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Reproducibility of Results
18.
Heliyon ; 5(12): e02899, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31872111

ABSTRACT

Cerebral cavernous malformations (CCMs) are characterized by abnormally dilated intracranial capillaries that result in increased susceptibility to stroke. Three genes have been identified as causes of CCMs; KRIT1 (CCM1), MGC4607 (CCM2) and PDCD10 (CCM3); one of them is disrupted in most CCM cases. It was demonstrated that both CCM1 and CCM3 bind to CCM2 to form a CCM signaling complex (CSC) to modulate angiogenesis. In this report, we deployed both RNA-seq and proteomic analysis of perturbed CSC after depletion of one of three CCM genes to generate interactomes for system-wide studies. Our results demonstrated a unique portrait detailing alterations in angiogenesis and vascular integrity. Interestingly, only in-direct overlapped alterations between RNA and protein levels were detected, supporting the existence of multiple layers of regulation in CSC cascades. Notably, this is the first report identifying that both ß4 integrin and CAV1 signaling are downstream of CSC, conveying the angiogenic signaling. Our results provide a global view of signal transduction modulated by the CSC, identifies novel regulatory signaling networks and key cellular factors associated with CSC.

19.
Sci Rep ; 9(1): 15808, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31676827

ABSTRACT

Cerebral cavernous malformations (CCMs) is a microvascular disorder in the central nervous system. Despite tremendous efforts, the causal genetic mutation in some CCM patients has not be identified, raising the possibility of an unknown CCM locus. The CCM2/MGC4607 gene has been identified as one of three known genes causing CCMs. In this report, we defined a total of 29 novel exons and 4 novel promoters in CCM2 genomic structure and subsequently identified a total of 50 new alternative spliced isoforms of CCM2 which eventually generated 22 novel protein isoforms. Genetic analysis of CCM2 isoforms revealed that the CCM2 isoforms can be classified into two groups based on their alternative promoters and alternative start codon exons. Our data demonstrated that CCM2 isoforms not only are specific in their subcellular compartmentation but also have distinct cellular expression patterns among various tissues and cells, indicating the pleiotropic cellular roles of CCM2 through their multiple isoforms. In fact, the complexity of the CCM2 genomic structure was reflected by the multiple layers of regulation of CCM2 expression patterns. At the transcriptional level, it is accomplished by alternative promoters, alternative splicing, and multiple transcriptional start sites and termination sites; while at the translational level, it is carried out with various cellular functions with a distinguishable CCM2 protein group pattern, specified abundance and composition of selective isoforms in a cell and tissue specific fashion. Through experimentation, we discovered a unique phosphotyrosine binding (PTB) domain, namely atypical phosphotyrosine binding (aPTB) domain. Some long CCM2 isoform proteins contain both classes of PTB domains, making them a dual PTB domain-containing protein. Both CCM1 and CCM3 can bind competitively to this aPTB domain, indicating CCM2 as the cornerstone for CCM signaling complex (CSC).


Subject(s)
Alternative Splicing , Carrier Proteins/genetics , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...