Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Int J Mol Sci ; 24(7)2023 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-37047353

RESUMO

Sigma-2 receptor (S2R) is a S2R ligand-binding site historically associated with reportedly 21.5 kDa proteins that have been linked to several diseases, such as cancer, Alzheimer's disease, and schizophrenia. The S2R is highly expressed in various tumors, where it correlates with the proliferative status of the malignant cells. Recently, S2R was reported to be the transmembrane protein TMEM97. Prior to that, we had been investigating the translocator protein (TSPO) as a potential 21.5 kDa S2R candidate protein with reported heme and sterol associations. Here, we investigate the contributions of TMEM97 and TSPO to S2R activity in MCF7 breast adenocarcinoma and MIA PaCa-2 (MP) pancreatic carcinoma cells. Additionally, the role of the reported S2R-interacting partner PGRMC1 was also elucidated. Proximity ligation assays and co-immunoprecipitation show a functional association between S2R and TSPO. Moreover, a close physical colocalization of TMEM97 and TSPO was found in MP cells. In MCF7 cells, co-immunoprecipitation only occurred with TMEM97 but not with PGRMC1, which was further confirmed by confocal microscopy experiments. Treatment with the TMEM97 ligand 20-(S)-hydroxycholesterol reduced co-immunoprecipitation of both TMEM97 and PGRMC1 in immune pellets of immunoprecipitated TSPO in MP cells. To the best of our knowledge, this is the first suggestion of a (functional) interaction between TSPO and TMEM97 that can be affected by S2R ligands.


Assuntos
Receptores sigma , Humanos , Ligantes , Ligação Proteica , Receptores sigma/metabolismo , Sítios de Ligação , Receptores de GABA/metabolismo , Proteínas de Membrana/metabolismo , Receptores de Progesterona/metabolismo
2.
Front Biosci (Landmark Ed) ; 27(11): 317, 2022 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-36472108

RESUMO

The title usage of Unde venisti 'from where have you come' is from a now dead language (Latin) that foundationally influenced modern English (not the major influence, but an essential formative one). This is an apt analogy for how both the ancient eukaryotic and eumetazoan functions of PGRMC proteins (PGRMC1 and PGRMC2 in mammals) probably influence modern human biology: via a formative trajectory from an evolutionarily foundational fulcrum. There is an arguable probability, although not a certainty, that PGRMC-like proteins were involved in eukaryogenesis. If so, then the proto-eukaryotic ancestral protein is modelled as having initiated the oxygen-induced and CYP450 (Cytochrome P450)-mediated synthesis of sterols in the endoplasmic reticulum to regulate proto-mitochondrial activity and heme homeostasis, as well as having enabled sterol transport between endoplasmic reticulum (ER) and mitochondria membranes involving the actin cytoskeleton, transport of heme from mitochondria, and possibly the regulation/origins of mitosis/meiosis. Later, during animal evolution, the last eumetazoan common ancestor (LEUMCA) acquired PGRMC phosphorylated tyrosines coincidentally with the gastrulation organizer, Netrin/deleted in colorectal carcinoma (DCC) signaling, muscle fibers, synapsed neurons, and neural recovery via a sleep-like process. Modern PGRMC proteins regulate multiple functions, including CYP450-mediated steroidogenesis, membrane trafficking, heme homeostasis, glycolysis/Warburg effect, fatty acid metabolism, mitochondrial regulation, and genomic CpG epigenetic regulation of gene expression. The latter imposes the system of differentiation status-sensitive cell-type specific proteomic complements in multi-tissued descendants of the LEUMCA. This paper attempts to trace PGRMC functions through time, proposing that key functions were involved in early eukaryotes, and were later added upon in the LEUMCA. An accompanying paper considers the implications of this awareness for human health and disease.


Assuntos
Eucariotos , Proteômica , Animais , Humanos , Epigênese Genética , Receptores de Progesterona/metabolismo , Glicólise , Heme/metabolismo , Mamíferos/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo
3.
Front Biosci (Landmark Ed) ; 27(11): 318, 2022 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-36472116

RESUMO

The title usage of Latin Quo vadis 'where are you going' extends the question Unde venisti from where 'did you come?' posed in the accompanying paper and extends consideration of how ancient eukaryotic and eumetazoan functions of progesterone receptor membrane component (PGRMC) proteins (PGRMC1 and PGRMC2 in mammals) could influence modern human health and disease. This paper attempts to extrapolate to modern biology in terms of extensions of hypothetical ancestral functional states from early eukaryotes and the last eumetazoan common ancestor (LEUMCA), to relativize human metabolic physiology and disease. As novel cell types and functional specializations appeared in bilaterian animals, PGRMC functions are hypothesized to have continued to be part of the toolkit used to develop new cell types and manage increasingly complex tasks such as nerve-gut-microbiome neuronal and hormonal communication. A critical role of PGRMC (as one component of a new eumetazoan genetic machinery) is proposed in LEUMCA endocrinology, neurogenesis, and nerve-gut communication with possible involvement in circadian nicotinamide adenine dinucleotide synthesis. This model would explain the contribution of PGRMC to metabolic and differentiation/behavioral changes observed in age-related diseases like diabetes, cancer and perhaps aging itself. Consistent with proposed key regulation of neurogenesis in the LEUMCA, it is argued that Alzheimer's disease is the modern pathology that most closely reflects the suite of functions related to PGRMC biology, with the 'usual suspect' pathologies possibly being downstream of PGRMC1. Hopefully, these thoughts help to signpost directions for future research.


Assuntos
Eucariotos , Receptores de Progesterona , Animais , Humanos , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Biologia , Mamíferos/metabolismo , Proteínas de Membrana/genética
4.
FEBS Lett ; 596(18): 2409-2417, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35993565

RESUMO

We recently reported that the membrane-associated progesterone receptor (MAPR) protein family (mammalian members: PGRMC1, PGRMC2, NEUFC and NENF) originated from a new class of prokaryotic cytochrome b5 (cytb5 ) domain proteins, called cytb5M (MAPR-like). Relative to classical cytb5 proteins, MAPR and ctyb5M proteins shared unique sequence elements and a distinct heme-binding orientation at an approximately 90° rotation relative to classical cytb5 , as demonstrated in the archetypal crystal structure of a cytb5M protein (PDB accession number 6NZX). Here, we present the crystal structure of an archaeal cytb5M domain (Methanococcoides burtonii WP_011499504.1, PDB:6VZ6). It exhibits similar heme binding to the 6NZX cytb5M , supporting the deduction that MAPR-like heme orientation was inherited from the prokaryotic ancestor of the original eukaryotic MAPR gene.


Assuntos
Citocromos b , Receptores de Progesterona , Animais , Archaea/genética , Archaea/metabolismo , Citocromos b/genética , Citocromos b/metabolismo , Citocromos b5/genética , Heme/metabolismo , Mamíferos , Ligação Proteica , Receptores de Progesterona/genética
5.
Cancers (Basel) ; 13(22)2021 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-34830790

RESUMO

In previous studies, we reported that progesterone receptor membrane component 1 (PGRMC1) is implicated in progestin signaling and possibly associated with increased breast cancer risk upon combined hormone replacement therapy. To gain mechanistic insight, we searched for potential PGRMC1 interaction partners upon progestin treatment by co-immunoprecipitation and mass spectrometry. The interactions with the identified partners were further characterized with respect to PGRMC1 phosphorylation status and with emphasis on the crosstalk between PGRMC1 and estrogen receptor α (ERα). We report that PGRMC1 overexpression resulted in increased proliferation of hormone receptor positive breast cancer cell lines upon treatment with a subgroup of progestins including norethisterone and dydrogesterone that promote PGRMC1-phosphorylation on S181. The ERα modulators prohibitin-1 (PHB1) and prohibitin-2 (PHB2) interact with PGRMC1 in dependency on S181-phosphorylation upon treatment with the same progestins. Moreover, increased interaction between PGRMC1 and PHBs correlated with decreased binding of PHBs to ERα and subsequent ERα activation. Inhibition of either PGRMC1 or ERα abolished this effect. In summary, we provide strong evidence that activated PGRMC1 associates with PHBs, competitively removing them from ERα, which then can develop its transcriptional activities on target genes. This study emphasizes the role of PGRMC1 in a key breast cancer signaling pathway which may provide a new avenue to target hormone-dependent breast cancer.

6.
Cancers (Basel) ; 13(3)2021 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-33572771

RESUMO

This is a preface by the guest editors of the special issue of Cancers featuring the biology of progesterone (P4) receptor membrane component (PGRMC) proteins as it relates to metabolism and cancer [...].

7.
eNeuro ; 7(6)2020.
Artigo em Inglês | MEDLINE | ID: mdl-33028631

RESUMO

The σ-2 receptor (S2R) complex has been implicated in CNS disorders ranging from anxiety and depression to neurodegenerative disorders such as Alzheimer's disease (AD). The proteins comprising the S2R complex impact processes including autophagy, cholesterol synthesis, progesterone signaling, lipid membrane-bound protein trafficking, and receptor stabilization at the cell surface. While there has been much progress in understanding the role of S2R in cellular processes and its potential therapeutic value, a great deal remains unknown. The International Symposium on Sigma-2 Receptors is held in conjunction with the annual Society for Neuroscience (SfN) conference to promote collaboration and advance the field of S2R research. This review summarizes updates presented at the Fourth International Symposium on Sigma-2 Receptors: Role in Health and Disease, a Satellite Symposium held at the 2019 SfN conference. Interdisciplinary members of the S2R research community presented both previously published and preliminary results from ongoing studies of the role of S2R in cellular metabolism, the anatomic and cellular expression patterns of S2R, the relationship between S2R and amyloid ß (Aß) in AD, the role of S2R complex protein PGRMC1 in health and disease, and the efforts to design new S2R ligands for the purposes of research and drug development. The proceedings from this symposium are reported here as an update on the field of S2R research, as well as to highlight the value of the symposia that occur yearly in conjunction with the SfN conference.


Assuntos
Doença de Alzheimer , Receptores sigma , Peptídeos beta-Amiloides , Humanos , Proteínas de Membrana , Progesterona , Receptores de Progesterona
9.
BMC Mol Cell Biol ; 21(1): 26, 2020 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-32293262

RESUMO

BACKGROUND: Progesterone receptor membrane component 1 (PGRMC1) is often elevated in cancers, and exists in alternative states of phosphorylation. A motif centered on PGRMC1 Y180 was evolutionarily acquired concurrently with the embryological gastrulation organizer that orchestrates vertebrate tissue differentiation. RESULTS: Here, we show that mutagenic manipulation of PGRMC1 phosphorylation alters cell metabolism, genomic stability, and CpG methylation. Each of several mutants elicited distinct patterns of genomic CpG methylation. Mutation of S57A/Y180/S181A led to increased net hypermethylation, reminiscent of embryonic stem cells. Pathways enrichment analysis suggested modulation of processes related to animal cell differentiation status and tissue identity, as well as cell cycle control and ATM/ATR DNA damage repair regulation. We detected different genomic mutation rates in culture. CONCLUSIONS: A companion manuscript shows that these cell states dramatically affect protein abundances, cell and mitochondrial morphology, and glycolytic metabolism. We propose that PGRMC1 phosphorylation status modulates cellular plasticity mechanisms relevant to early embryological tissue differentiation.


Assuntos
Fosforilação , Receptores de Progesterona , Animais , Diferenciação Celular , Linhagem Celular , Metilação de DNA , Doença , Embriologia , Epigenômica , Humanos , Proteínas de Membrana/biossíntese , Proteínas de Membrana/metabolismo , Camundongos , Mutação , Taxa de Mutação , Processamento de Proteína Pós-Traducional , Receptores de Progesterona/biossíntese , Receptores de Progesterona/metabolismo
10.
Mol Phylogenet Evol ; 148: 106814, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32278076

RESUMO

The membrane-associated progesterone receptor (MAPR) family consists of heme-binding proteins containing a cytochrome b5 (cytb5) domain characterized by the presence of a MAPR-specific interhelical insert region (MIHIR) between helices 3 and 4 of the canonical cytb5-domain fold. Animals possess three MAPR genes (PGRMC-like, Neuferricin and Neudesin). Here we show that all three animal MAPR genes were already present in the common ancestor of the opisthokonts (comprising animals and fungi as well as related single-celled taxa). All three MAPR genes acquired extensions C-terminal to the cytb5 domain, either before or with the evolution of animals. The archetypical MAPR protein, progesterone receptor membrane component 1 (PGRMC1), contains phosphorylated tyrosines Y139 and Y180. The combination of Y139/Y180 appeared in the common ancestor of cnidarians and bilaterians, along with an early embryological organizer and synapsed neurons, and is strongly conserved in all bilaterian animals. A predicted protein interaction motif in the PGRMC1 MIHIR is potentially regulated by Y139 phosphorylation. A multilayered model of animal MAPR function acquisition includes some pre-metazoan functions (e.g., heme binding and cytochrome P450 interactions) and some acquired animal-specific functions that involve regulation of strongly conserved protein interaction motifs acquired by animals (Metazoa). This study provides a conceptual framework for future studies, against which especially PGRMC1's multiple functions can perhaps be stratified and functionally dissected.


Assuntos
Eucariotos/metabolismo , Proteínas de Membrana/metabolismo , Receptores de Progesterona/metabolismo , Sequência de Aminoácidos , Animais , Evolução Molecular , Proteínas de Membrana/química , Filogenia , Ligação Proteica , Domínios Proteicos , Receptores de Progesterona/química , Receptores de Progesterona/genética
11.
BMC Mol Cell Biol ; 21(1): 24, 2020 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-32245408

RESUMO

BACKGROUND: Progesterone Receptor Membrane Component 1 (PGRMC1) is expressed in many cancer cells, where it is associated with detrimental patient outcomes. It contains phosphorylated tyrosines which evolutionarily preceded deuterostome gastrulation and tissue differentiation mechanisms. RESULTS: We demonstrate that manipulating PGRMC1 phosphorylation status in MIA PaCa-2 (MP) cells imposes broad pleiotropic effects. Relative to parental cells over-expressing hemagglutinin-tagged wild-type (WT) PGRMC1-HA, cells expressing a PGRMC1-HA-S57A/S181A double mutant (DM) exhibited reduced levels of proteins involved in energy metabolism and mitochondrial function, and altered glucose metabolism suggesting modulation of the Warburg effect. This was associated with increased PI3K/AKT activity, altered cell shape, actin cytoskeleton, motility, and mitochondrial properties. An S57A/Y180F/S181A triple mutant (TM) indicated the involvement of Y180 in PI3K/AKT activation. Mutation of Y180F strongly attenuated subcutaneous xenograft tumor growth in NOD-SCID gamma mice. Elsewhere we demonstrate altered metabolism, mutation incidence, and epigenetic status in these cells. CONCLUSIONS: Altogether, these results indicate that mutational manipulation of PGRMC1 phosphorylation status exerts broad pleiotropic effects relevant to cancer and other cell biology.


Assuntos
Fosforilação , Receptores de Progesterona , Animais , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Forma Celular , Metabolismo Energético , Glicólise , Humanos , Proteínas de Membrana/biossíntese , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Mitocôndrias/metabolismo , Neoplasias , Fosfatidilinositol 3-Quinases/metabolismo , Receptores de Progesterona/biossíntese , Receptores de Progesterona/metabolismo
12.
Biochem Biophys Res Commun ; 524(1): 64-69, 2020 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-31980178

RESUMO

PGRMC1 is a protein from the MAPR family with a range of cellular functions. PGRMC1 has been described to play a role in fertility, neuroprotection, steroidogenesis, membrane trafficking and in cancer cell biology. PGRMC1 represents a likely key regulator of cell metabolism and proliferation, as well as a potential target for anti-cancer therapies. To further understand the functions of PGRMC1 and the mechanism of the small molecule inhibitor of PGRMC1, AG-205, proteins differentially bound to PGRMC1 were identified following AG-205 treatment of MIA PaCa-2 cells. Our results suggest that AG-205 influences PGRMC1 interactions with the actin cytoskeleton. The binding of two PGRMC1-associated proteins that support this, RACK1 and alpha-Actinin-1, was reduced following AG-205 treatment. The biology associated with PGRMC1 binding partners identified here merits further investigation.


Assuntos
Actinas/metabolismo , Indóis/metabolismo , Proteínas de Membrana/antagonistas & inibidores , Receptores de Progesterona/antagonistas & inibidores , Citoesqueleto de Actina/metabolismo , Linhagem Celular Tumoral , Humanos , Espectrometria de Massas , Ligação Proteica , Receptores de Quinase C Ativada/metabolismo
13.
Diagn Cytopathol ; 46(11): 927-935, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30284391

RESUMO

BACKGROUND: The cytomorphological features in the distinction between phyllodes tumour (PT) and fibroadenoma (FA) on fine needle aspiration biopsy (FNAB) remains challenging because of the biphasic nature of these lesions and the rarity of PT. METHODS: FNAB smears of histologically confirmed PT (N = 26) and FA (N = 78) cases were retrieved from a retrospective database interrogation from the Department of Cytology/Tissue Pathology, ICPMR Pathology West (Cerner Millennium) in Westmead Hospital. For each case, two smears were selected, de-identified and independently reviewed by four observers comprising two cytologists and two cytopathologists. Cytological parameters examined included detailed evaluation of smear cellularity, epithelial and stromal components as well as the smear background. RESULTS: The cytological features of moderate to marked stromal cellularity and stromal nuclear atypia were more evident in PT than in FA, identified by three out of four observers. The epithelial characteristics, presence of macrophages, multinucleated giant cells and blood vessels showed no statistically significant differences in the distinction between the two lesions. CONCLUSION: The results of this study indicate that in all of the cytological features assessed for PT and FA, no single cytological feature was found to be statistically significant in identifying PT across all observers. This reflects the overlap of cytological features seen in these lesions. FNAB cytology cannot reliably distinguish FA and PT.


Assuntos
Neoplasias da Mama/patologia , Fibroadenoma/patologia , Tumor Filoide/patologia , Adolescente , Adulto , Biópsia por Agulha Fina/normas , Feminino , Humanos , Pessoa de Meia-Idade , Variações Dependentes do Observador
14.
Biosci Trends ; 11(2): 179-192, 2017 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-28250339

RESUMO

A complex PGRMC1-centred regulatory system controls multiple cell functions. Although PGRMC1 is phosphorylated at several positions, we do not understand the mechanisms regulating its function. PGRMC1 is the archetypal member of the membrane associated progesterone receptor (MAPR) family. Phylogentic comparison of MAPR proteins suggests that the ancestral metazoan "PGRMC-like" MAPR gene resembled PGRMC1/PGRMC2, containing the equivalents of PGRMC1 Y139 and Y180 SH2 target motifs. It later acquired a CK2 site with phosphoacceptor at S181. Separate PGRMC1 and PGRMC2 genes with this "PGRMC-like" structure diverged after the separation of vertebrates from protochordates. Terrestrial tetrapods possess a novel proline-rich PGRMC1 SH3 target motif centred on P64 which in mammals is augmented by a phosphoacceptor at PGRMC1 S54, and in primates by an additional S57 CK2 site. All of these phosphoacceptors are phosphorylated in vivo. This study suggests that an increasingly sophisticated system of PGRMC1-modulated multicellular functional regulation has characterised animal evolution since Precambrian times.


Assuntos
Proteínas de Membrana/metabolismo , Receptores de Progesterona/metabolismo , Animais , Humanos , Proteínas de Membrana/genética , Receptores de Progesterona/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
15.
J Steroid Biochem Mol Biol ; 171: 11-33, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28104494

RESUMO

Progesterone Receptor Membrane Component 1 (PGRMC1) is located in many different subcellular locations with many different attested and probably location-specific functions. PGRMC1 was recently identified in the mitochondrial outer membrane where it interacts with ferrochelatase, the last enzyme in the heme synthetic pathway. It has been proposed that PGRMC1 may act as a chaperone to shuttle newly synthesized heme from the mitochondrion to cytochrome P450 (cyP450) enzymes. Here we consider potential roles that PGRMC1 may play in transferring heme, and other small hydrophobic ligands such as cholesterol and steroids, between the hydrophobic compartment of the membrane lipid bilayer interior to aqueous proteins, and perhaps to the membranes of other organelles. We review the synthesis and roles of especially PGRMC1- and cyP450-bound heme, the sources and transport of cholesterol, the involvement of PGRMC1 in cholesterol regulation, and the production of the first progestogen pregnenolone from cholesterol. We also show by clustering by inferred models of evolution (CLIME) analysis that PGRMC1 and related proteins exhibit co-evolution with a series of cyP450 enzymes, as well as a group of mitochondrial proteins lacking in several parasitic protist groups. Altogether, PGRMC1 is implicated with important roles in sterol synthesis and energy regulation that are dispensable in certain parasites. Some novel hypothetical models for PGRMC1 function are proposed to direct future investigative research.


Assuntos
Proteínas de Membrana/metabolismo , Modelos Moleculares , Receptores de Progesterona/metabolismo , Sequência de Aminoácidos , Animais , Transporte Biológico , Colesterol/química , Colesterol/metabolismo , Sequência Conservada , Metabolismo Energético , Evolução Molecular , Humanos , Interações Hidrofóbicas e Hidrofílicas , Ligantes , Proteínas de Membrana/química , Proteínas de Membrana/genética , Filogenia , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Receptores de Progesterona/química , Receptores de Progesterona/genética , Homologia de Sequência de Aminoácidos , Esteroides/química , Esteroides/metabolismo
16.
Oncotarget ; 7(32): 50822-50827, 2016 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-27448967

RESUMO

Progesterone receptor membrane component 1 (PGRMC1) is a multifunctional protein implicated in multiple pathologies, including cancer and Alzheimer's disease. The recently published structure of PGRMC1 revealed heme-mediated dimerization that directed the PGRMC1-dependent cytochrome P450-mediated detoxification of doxorubicin. We describe here how the PGRMC1 structure also enables important new insights into the possible regulation of PGRMC1 function by phosphorylation. Predicted regulatory interaction sites for SH2- and SH3-domain proteins are in non-structured regions that could be available to cytoplasmic enzymes. Further to the published interpretation, we suggest that phosphorylation of PGRMC1 at position Y113 may promote the attested membrane trafficking function of PGRMC1. To stimulate further experimentation, we also discuss that heme-mediated dimerization of PGRMC1 and membrane trafficking may be mutually exclusive functions. These roles could potentially be reciprocally regulated by phosphorylation/dephosphorylation at Y113. It follows that the phosphorylation status of PGRMC1 should be further explored in order to better understand many of its proposed biological functions.


Assuntos
Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Receptores de Progesterona/química , Receptores de Progesterona/metabolismo , Humanos , Fosforilação , Transporte Proteico/fisiologia
17.
Biochim Biophys Acta ; 1866(2): 339-349, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27452206

RESUMO

Progesterone receptor membrane component 1 (PGRMC1) is a multi-functional protein with a heme-binding moiety related to that of cytochrome b5, which is a putative progesterone receptor. The recently solved PGRMC1 structure revealed that heme-binding involves coordination by a tyrosinate ion at Y113, and induces dimerization which is stabilized by hydrophobic stacking of heme on adjacent monomers. Dimerization is required for association with cytochrome P450 (cyP450) enzymes, which mediates chemoresistance to doxorubicin and may be responsible for PGRMC1's anti-apoptotic activity. Here we review the multiple attested involvement of PGRMC1 in diverse functions, including regulation of cytochrome P450, steroidogenesis, vesicle trafficking, progesterone signaling and mitotic spindle and cell cycle regulation. Its wide range of biological functions is attested to particularly by its emerging association with cancer and progesterone-responsive female reproductive tissues. PGRMC1 exhibits all the hallmarks of a higher order nexus signal integration hub protein. It appears capable of acting as a detector that integrates information from kinase/phosphatase pathways with heme and CO levels and probably redox status.


Assuntos
Proteínas de Membrana/fisiologia , Neoplasias/metabolismo , Receptores de Progesterona/fisiologia , Ciclo Celular , Proliferação de Células , Humanos , Proteínas de Membrana/química , Neoplasias/patologia , Multimerização Proteica , Receptores de Progesterona/química , Receptores sigma/fisiologia
18.
Sci Rep ; 6: 23453, 2016 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-27029742

RESUMO

Automated and unbiased methods of non-invasive cell monitoring able to deal with complex biological heterogeneity are fundamentally important for biology and medicine. Label-free cell imaging provides information about endogenous autofluorescent metabolites, enzymes and cofactors in cells. However extracting high content information from autofluorescence imaging has been hitherto impossible. Here, we quantitatively characterise cell populations in different tissue types, live or fixed, by using novel image processing and a simple multispectral upgrade of a wide-field fluorescence microscope. Our optimal discrimination approach enables statistical hypothesis testing and intuitive visualisations where previously undetectable differences become clearly apparent. Label-free classifications are validated by the analysis of Classification Determinant (CD) antigen expression. The versatility of our method is illustrated by detecting genetic mutations in cancer, non-invasive monitoring of CD90 expression, label-free tracking of stem cell differentiation, identifying stem cell subpopulations with varying functional characteristics, tissue diagnostics in diabetes, and assessing the condition of preimplantation embryos.


Assuntos
Rastreamento de Células/métodos , Diabetes Mellitus Experimental/metabolismo , Mutação , Imagem Óptica/métodos , Neoplasias Pancreáticas/ultraestrutura , Antígenos Thy-1/genética , Animais , Blastocisto/metabolismo , Blastocisto/ultraestrutura , Diferenciação Celular , Linhagem Celular Tumoral , Rastreamento de Células/instrumentação , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Expressão Gênica , Regulação da Expressão Gênica , Humanos , Processamento de Imagem Assistida por Computador/estatística & dados numéricos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Imagem Óptica/estatística & dados numéricos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Antígenos Thy-1/metabolismo
19.
PLoS One ; 9(11): e111899, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25390692

RESUMO

Amyloid beta (Abeta) 1-42 oligomers accumulate in brains of patients with Mild Cognitive Impairment (MCI) and disrupt synaptic plasticity processes that underlie memory formation. Synaptic binding of Abeta oligomers to several putative receptor proteins is reported to inhibit long-term potentiation, affect membrane trafficking and induce reversible spine loss in neurons, leading to impaired cognitive performance and ultimately to anterograde amnesia in the early stages of Alzheimer's disease (AD). We have identified a receptor not previously associated with AD that mediates the binding of Abeta oligomers to neurons, and describe novel therapeutic antagonists of this receptor capable of blocking Abeta toxic effects on synapses in vitro and cognitive deficits in vivo. Knockdown of sigma-2/PGRMC1 (progesterone receptor membrane component 1) protein expression in vitro using siRNA results in a highly correlated reduction in binding of exogenous Abeta oligomers to neurons of more than 90%. Expression of sigma-2/PGRMC1 is upregulated in vitro by treatment with Abeta oligomers, and is dysregulated in Alzheimer's disease patients' brain compared to age-matched, normal individuals. Specific, high affinity small molecule receptor antagonists and antibodies raised against specific regions on this receptor can displace synthetic Abeta oligomer binding to synaptic puncta in vitro and displace endogenous human AD patient oligomers from brain tissue sections in a dose-dependent manner. These receptor antagonists prevent and reverse the effects of Abeta oligomers on membrane trafficking and synapse loss in vitro and cognitive deficits in AD mouse models. These findings suggest sigma-2/PGRMC1 receptors mediate saturable oligomer binding to synaptic puncta on neurons and that brain penetrant, small molecules can displace endogenous and synthetic oligomers and improve cognitive deficits in AD models. We propose that sigma-2/PGRMC1 is a key mediator of the pathological effects of Abeta oligomers in AD and is a tractable target for small molecule disease-modifying therapeutics.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/química , Proteínas de Membrana/metabolismo , Fragmentos de Peptídeos/química , Receptores de Progesterona/metabolismo , Sinapses/efeitos dos fármacos , Doença de Alzheimer/metabolismo , Animais , Autorradiografia , Encéfalo/metabolismo , Membrana Celular/metabolismo , Cognição/efeitos dos fármacos , Transtornos Cognitivos/tratamento farmacológico , Humanos , Proteínas de Membrana/genética , Camundongos , Neurônios/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Progesterona/genética , Sinapses/metabolismo
20.
J Biomed Opt ; 19(8): 086016, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25140884

RESUMO

Measurement of endogenous free and bound NAD(P)H relative concentrations in living cells isa useful method for monitoring aspects of cellular metabolism, because the NADH∕NAD⁺ reduction-oxidation pair is crucial for electron transfer through the mitochondrial electron transport chain. Variations of free and bound NAD(P)H ratio are also implicated in cellular bioenergetic and biosynthetic metabolic changes accompanying cancer. This study uses two-photon fluorescence lifetime imaging microscopy (FLIM) to investigate metabolic changes in MCF10A premalignant breast cancer cells treated with a range of glycolysis inhibitors: namely, 2 deoxy-D-glucose, oxythiamine, lonidamine, and 4-(chloromethyl) benzoyl chloride, as well as the mitochondrial membrane uncoupling agent carbonyl cyanide m-chlorophenylhydrazone. Through systematic analysis of FLIM data from control and treated cancer cells, we observed that all glycolytic inhibitors apart from lonidamine had a slightly decreased metabolic rate and that the presence of serum in the culture medium generally marginally protected cells from the effect of inhibitors. Direct production of glycolytic L-lactate was also measured in both treated and control cells. The combination of these two techniques gave valuable insights into cell metabolism and indicated that FLIM was more sensitive than traditional biochemical methods, as it directly measured metabolic changes within cells as compared to quantification of lactate secreted by metabolically active cells.


Assuntos
Neoplasias da Mama/enzimologia , Inibidores Enzimáticos/farmacologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Microscopia de Fluorescência por Excitação Multifotônica/métodos , NAD/antagonistas & inibidores , NAD/metabolismo , Linhagem Celular Tumoral , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA