Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
JACC Case Rep ; 4(22): 1515-1521, 2022 Nov 16.
Article in English | MEDLINE | ID: mdl-36444176

ABSTRACT

Pericardial decompression syndrome (PDS) is a potentially fatal disorder of left ventricular function that sometimes occurs after drainage of a pericardial effusion for cardiac tamponade. Patients at risk for PDS are difficult to identify. Here, we report 2 cases where PDS developed after drainage of effusions that had been present for years, suggesting that patients with chronic effusions are at higher risk for PDS. (Level of Difficulty: Advanced.).

3.
Stem Cell Reports ; 8(4): 1086-1100, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28410642

ABSTRACT

Large-scale collections of induced pluripotent stem cells (iPSCs) could serve as powerful model systems for examining how genetic variation affects biology and disease. Here we describe the iPSCORE resource: a collection of systematically derived and characterized iPSC lines from 222 ethnically diverse individuals that allows for both familial and association-based genetic studies. iPSCORE lines are pluripotent with high genomic integrity (no or low numbers of somatic copy-number variants) as determined using high-throughput RNA-sequencing and genotyping arrays, respectively. Using iPSCs from a family of individuals, we show that iPSC-derived cardiomyocytes demonstrate gene expression patterns that cluster by genetic background, and can be used to examine variants associated with physiological and disease phenotypes. The iPSCORE collection contains representative individuals for risk and non-risk alleles for 95% of SNPs associated with human phenotypes through genome-wide association studies. Our study demonstrates the utility of iPSCORE for examining how genetic variants influence molecular and physiological traits in iPSCs and derived cell lines.


Subject(s)
Arrhythmias, Cardiac/genetics , Databases, Factual , Genetic Association Studies , Genetic Variation , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Arrhythmias, Cardiac/ethnology , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , Cell Differentiation , Cell Line , Cellular Reprogramming/genetics , Genotype , High-Throughput Nucleotide Sequencing , Humans , Induced Pluripotent Stem Cells/cytology , Multigene Family , Myocytes, Cardiac/cytology , Oligonucleotide Array Sequence Analysis , Phenotype , Polymorphism, Single Nucleotide , Racial Groups
4.
Pacing Clin Electrophysiol ; 39(11): 1254-1260, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27550834

ABSTRACT

BACKGROUND: Radiofrequency (RF) technology has improved detection of retained surgical sponges with a reported 100% sensitivity and specificity. However, the potential for interactions of the RF signals emitted by the detection system with cardiac implantable electronic devices (CIEDs) or temporary pacemakers may limit its use in those patients with these devices. This study investigated whether RF detection technology causes interference or clinically significant changes in the programmed settings of implanted pacemakers and defibrillators or temporary epicardial pacemakers. METHODS: Fifty patients who were scheduled either for CIED removal or placement of a temporary epicardial pacemaker (at the time of open heart surgery) were recruited for this study. Device settings and measurements from separate interrogations before and after scanning with the RF detection system were compared. For the temporary pacemakers, we observed for any changes in hemodynamics or signs of pacing interference. RESULTS: Twenty (40%) pacemakers, 20 (40%) implantable cardioverter defibrillators, and 10 (20%) temporary pacemakers were analyzed in this study. During scanning, no signal interference was detected in any permanent device, and there were no significant changes in programmed settings after scanning with the RF detection system. However, pacing inhibition was detected with temporary pacing systems when programmed to a synchronous mode (DDD). CONCLUSIONS: RF detection technology can be safely used to scan for retained surgical sponges in patients with permanent CIEDs and temporary pacemakers set to asynchronous mode.


Subject(s)
Defibrillators, Implantable , Foreign Bodies/diagnosis , Pacemaker, Artificial , Radio Waves , Aged , Female , Humans , Male , Middle Aged
5.
Genes (Basel) ; 6(2): 252-66, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25988259

ABSTRACT

The initiation of DNA replication is tightly regulated in order to ensure that the genome duplicates only once per cell cycle. In vertebrate cells, the unstable regulatory protein Geminin prevents a second round of DNA replication by inhibiting the essential replication factor Cdt1. Cdt1 recruits mini-chromosome maintenance complex (MCM2-7), the replication helicase, into the pre-replication complex (pre-RC) at origins of DNA replication. The mechanism by which Geminin inhibits MCM2-7 loading by Cdt1 is incompletely understood. The conventional model is that Geminin sterically hinders a direct physical interaction between Cdt1 and MCM2-7. Here, we describe an inactive missense mutant of Geminin, GemininAWA, which binds to Cdt1 with normal affinity yet is completely inactive as a replication inhibitor even when added in vast excess. In fact, GemininAWA can compete with GemininWT for binding to Cdt1 and prevent it from inhibiting DNA replication. GemininAWA does not inhibit the loading of MCM2-7 onto DNA in vivo, and in the presence of GemininAWA, nuclear DNA is massively over-replicated within a single S phase. We conclude that Geminin does not inhibit MCM loading by simple steric interference with a Cdt1-MCM2-7 interaction but instead works by a non-steric mechanism, possibly by inhibiting the histone acetyltransferase HBO1.

8.
Europace ; 16(3): 372-7, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24127355

ABSTRACT

AIMS: Managing an infection of the pocket of a cardiac implantable electronic device (CIED) is frequently challenging. The wound is often treated with a drain or wet-to-dry dressings that allow healing by secondary intention. Such treatment can prolong the hospital stay and can frequently result in a disfiguring scar. Negative pressure wound therapy (NPWT) has been frequently used to promote the healing of chronic or infected surgical wounds. Here we describe the first series of 28 patients in which NPWT was successfully used to treat CIED pocket infections. METHODS AND RESULTS: After removal of the CIED and debridement of the pocket, a negative pressure of 125 mmHg continuously applied to the wound through an occlusive dressing. Negative pressure wound therapy was continued for a median of 5 days (range 2-15 days) and drained an average of 260 mL sero-sanguineous fluid (range 35-970 mL). At the conclusion of NPWT, delayed primary closure of the pocket was performed with 1-0 prolene mattress sutures. The median length of stay after CIED extraction was 11.0 days (range 2-43 days). Virtually all infected pockets healed without complications and without evidence of recurrent infection over a median follow-up of 49 days (range 10-752 days). One patient developed a recurrent infection when NPWT was discontinued prematurely and a new device was implanted at the infected site. CONCLUSION: We conclude that NPWT is a safe and effective means to promote healing of infected pockets with a low incidence of recurrent infection and a satisfactory cosmetic result.


Subject(s)
Defibrillators, Implantable/adverse effects , Electrodes, Implanted/adverse effects , Negative-Pressure Wound Therapy/methods , Pacemaker, Artificial/adverse effects , Prosthesis-Related Infections/etiology , Prosthesis-Related Infections/therapy , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Negative-Pressure Wound Therapy/instrumentation , Occlusive Dressings , Surgical Wound Infection/etiology , Surgical Wound Infection/therapy , Treatment Outcome
10.
Dev Biol ; 371(1): 35-46, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22898305

ABSTRACT

Spermatogonial stem cells divide throughout life, maintaining their own population and giving rise to differentiated gametes. The unstable regulatory protein Geminin is thought to be one of the factors that determine whether stem cells continue to divide or terminally differentiate. Geminin regulates the extent of DNA replication and is thought to maintain cells in an undifferentiated state by inhibiting various transcription factors and chromatin remodeling proteins. To examine how Geminin might regulate spermatogenesis, we developed two conditional mouse models in which the Geminin gene (Gmnn) is deleted from either spermatogonia or meiotic spermatocytes. Deleting Geminin from spermatogonia causes complete sterility in male mice. Gmnn(-/-) spermatogonia disappear during the initial wave of mitotic proliferation that occurs during the first week of life. Gmnn(-/-) spermatogonia exhibit more double-stranded DNA breaks than control cells, consistent with a defect in DNA replication. They maintain expression of genes associated with the undifferentiated state and do not prematurely express genes characteristic of more differentiated spermatogonia. In contrast, deleting Geminin from spermatocytes does not disrupt meiosis or the differentiation of spermatids into mature sperm. In females, Geminin is not required for meiosis, oocyte differentiation, or fertility after the embryonic period of mitotic proliferation has ceased. We conclude that Geminin is absolutely required for mitotic proliferation of spermatogonia but does not regulate their differentiation. Our results suggest that Geminin maintains replication fidelity during the mitotic phase of spermatogenesis, ensuring the precise duplication of genetic information for transmission to the next generation.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Proliferation , Fertility/genetics , Mitosis/physiology , Nuclear Proteins/metabolism , Spermatogonia/physiology , Animals , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/genetics , DNA Breaks, Double-Stranded , DNA Replication/genetics , Galactosides , Geminin , Gene Knockout Techniques , Immunohistochemistry , Indoles , Male , Mice , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Real-Time Polymerase Chain Reaction , Spermatogonia/cytology
11.
PLoS One ; 7(5): e38009, 2012.
Article in English | MEDLINE | ID: mdl-22662261

ABSTRACT

In many organisms early development is under control of the maternal genome and zygotic gene expression is delayed until the mid-blastula transition (MBT). As zygotic transcription initiates, cell cycle checkpoints become activated and the tempo of cell division slows. The mechanisms that activate zygotic transcription at the MBT are incompletely understood, but they are of interest because they may resemble mechanisms that cause stem cells to stop dividing and terminally differentiate. The unstable regulatory protein Geminin is thought to coordinate cell division with cell differentiation. Geminin is a bi-functional protein. It prevents a second round of DNA replication during S and G2 phase by binding and inhibiting the essential replication factor Cdt1. Geminin also binds and inhibits a number of transcription factors and chromatin remodeling proteins and is thought to keep dividing cells in an undifferentiated state. We previously found that the cells of Geminin-deficient Xenopus embryos arrest in G2 phase just after the MBT then disintegrate at the onset of gastrulation. Here we report that they also fail to express most zygotic genes. The gene expression defect is cell-autonomous and is reproduced by over-expressing Cdt1 or by incubating the embryos in hydroxyurea. Geminin deficient and hydroxyurea-treated blastomeres accumulate DNA damage in the form of double stranded breaks. Bypassing the Chk1 pathway overcomes the cell cycle arrest caused by Geminin depletion but does not restore zygotic gene expression. In fact, bypassing the Chk1 pathway by itself induces double stranded breaks and abolishes zygotic transcription. We did not find evidence that Geminin has a replication-independent effect on transcription. We conclude that Geminin is required to maintain genome integrity during the rapid cleavage divisions, and that DNA damage disrupts zygotic gene transcription at the MBT, probably through activation of DNA damage checkpoint pathways.


Subject(s)
Blastula/metabolism , Cell Cycle Proteins/genetics , Gene Expression Regulation, Developmental , Xenopus/embryology , Xenopus/genetics , Zygote/metabolism , Animals , Cell Cycle Checkpoints , Cell Cycle Proteins/metabolism , DNA Damage , DNA-Binding Proteins/metabolism , Female , Geminin , Gene Deletion , Hydroxyurea/pharmacology , Male , Mutation , Protein Binding , T-Box Domain Proteins/metabolism , Transcription, Genetic/drug effects , Xenopus Proteins/metabolism
13.
PLoS One ; 6(3): e17736, 2011 Mar 09.
Article in English | MEDLINE | ID: mdl-21408022

ABSTRACT

Neural stem cells (NSCs) are the progenitors of neurons and glial cells during both embryonic development and adult life. The unstable regulatory protein Geminin (Gmnn) is thought to maintain neural stem cells in an undifferentiated state while they proliferate. Geminin inhibits neuronal differentiation in cultured cells by antagonizing interactions between the chromatin remodeling protein Brg1 and the neural-specific transcription factors Neurogenin and NeuroD. Geminin is widely expressed in the CNS during throughout embryonic development, and Geminin expression is down-regulated when neuronal precursor cells undergo terminal differentiation. Over-expression of Geminin in gastrula-stage Xenopus embryos can expand the size of the neural plate. The role of Geminin in regulating vertebrate neurogenesis in vivo has not been rigorously examined. To address this question, we created a strain of Nestin-Cre/Gmnn(fl/fl) mice in which the Geminin gene was specifically deleted from NSCs. Interestingly, we found no major defects in the development or function of the central nervous system. Neural-specific Gmnn(Δ/Δ) mice are viable and fertile and display no obvious neurological or neuroanatomical abnormalities. They have normal numbers of BrdU(+) NSCs in the subgranular zone of the dentate gyrus, and Gmnn(Δ/Δ) NSCs give rise to normal numbers of mature neurons in pulse-chase experiments. Gmnn(Δ/Δ) neurosphere cells differentiate normally into both neurons and glial cells when grown in growth factor-deficient medium. Both the growth rate and the cell cycle distribution of cultured Gmnn(Δ/Δ) neurosphere cells are indistinguishable from controls. We conclude that Geminin is largely dispensable for most of embryonic and adult mammalian neurogenesis.


Subject(s)
Cell Division , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis , Nuclear Proteins/deficiency , Animals , Cell Aggregation , Cell Cycle , Cell Cycle Proteins/metabolism , Cell Differentiation , DNA Replication , Female , Fertility , Geminin , Gene Deletion , Hippocampus/cytology , Hippocampus/pathology , Integrases/metabolism , Intermediate Filament Proteins/metabolism , Kinetics , Male , Mice , Nerve Tissue Proteins/metabolism , Nestin , Nuclear Proteins/metabolism , Survival Analysis
14.
J Clin Invest ; 120(12): 4303-15, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21041951

ABSTRACT

HSCs maintain the circulating blood cell population. Defects in the orderly pattern of hematopoietic cell division and differentiation can lead to leukemia, myeloproliferative disorders, or marrow failure; however, the factors that control this pattern are incompletely understood. Geminin is an unstable regulatory protein that regulates the extent of DNA replication and is thought to coordinate cell division with cell differentiation. Here, we set out to determine the function of Geminin in hematopoiesis by deleting the Geminin gene (Gmnn) from mouse bone marrow cells. This severely perturbed the pattern of blood cell production in all 3 hematopoietic lineages (erythrocyte, megakaryocyte, and leukocyte). Red cell production was virtually abolished, while megakaryocyte production was greatly enhanced. Leukocyte production transiently decreased and then recovered. Stem and progenitor cell numbers were preserved, and Gmnn(­/­) HSCs successfully reconstituted hematopoiesis in irradiated mice. CD34(+) Gmnn(­/­) leukocyte precursors displayed DNA overreplication and formed extremely small granulocyte and monocyte colonies in methylcellulose. While cultured Gmnn(­/­) mega-karyocyte-erythrocyte precursors did not form erythroid colonies, they did form greater than normal numbers of megakaryocyte colonies. Gmnn(­/­) megakaryocytes and erythroblasts had normal DNA content. These data led us to postulate that Geminin regulates the relative production of erythrocytes and megakaryocytes from megakaryocyte-erythrocyte precursors by a replication-independent mechanism.


Subject(s)
Anemia/genetics , Hematopoietic Stem Cells/metabolism , Nuclear Proteins/deficiency , Thrombocytosis/genetics , Anemia/blood , Animals , Base Sequence , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Colony-Forming Units Assay , DNA Primers/genetics , DNA Replication , Erythroblasts/metabolism , Erythroblasts/pathology , Female , Geminin , Gene Expression Regulation, Developmental , Gene Targeting , Hematopoiesis/genetics , Hematopoiesis/physiology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/pathology , In Vitro Techniques , Male , Megakaryocytes/metabolism , Megakaryocytes/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Thrombocytosis/blood
15.
J Hematop ; 3(1): 41-6, 2010 May 05.
Article in English | MEDLINE | ID: mdl-21544187

ABSTRACT

Rapidly progressive heart failure is commonly caused by an extensive myocardial infarction, a mechanical complication of infarction, myocarditis, or acute valvular insufficiency. We present an unusual case that was caused by a diffuse infiltration of the myocardium with leukemic cells (myeloid sarcoma). The patient presented with episodic shortness of breath, he was anemic and thrombocytopenic, and his bone marrow biopsy revealed myelodysplastic syndrome from treatment for oligodendroglioma. His clinical course was characterized by a chronic leak of cardiac enzymes, a new right bundle branch block, and a large pericardial effusion causing tamponade and death from fulminant heart failure and ventricular arrhythmias within 2 weeks. At autopsy, the heart was massively infiltrated with myeloblasts and other immature myeloid cells. There was no evidence of acute leukemia in the bone marrow or peripheral blood. Cardiac infiltration in a patient with myelodysplastic syndrome is extremely rare, especially in the absence of bone marrow involvement by blasts. The recognition of this entity is becoming increasingly important as the incidence of cardiac myeloid sarcoma may be on the rise as the number of patients receiving chemotherapy increases.

16.
Dev Dyn ; 238(10): 2614-21, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19705441

ABSTRACT

The cell nucleus must be inactivated or destroyed in order to generate feeder layers for cultured cells or to prepare recipient egg cells for nuclear transfer. Existing enucleation techniques are either cumbersome or employ toxic chemicals. Here we report a new method to enucleate cells by treatment with a psoralen and long-wave ultraviolet light. The technique is >90% efficient and causes little cytoplasmic damage to the treated cell. We have used psoralen treatment to enucleate a wide variety of cells, including eggs, sperm, HeLa cells, and fibroblasts. Colonies of human embryonic stem cells (hESCs) and human keratinocyte precursors grown on psoralen-treated feeders are indistinguishable from those grown on gamma-irradiated or mitomycin C-treated cells. Psoralen enucleation provides a rapid, simple, and non-toxic method to generate feeder cells. The technique is also useful for nuclear transfer studies in species with large eggs whose cleavage divisions are not regulated by cell-cycle checkpoints.


Subject(s)
Cell Nucleus , Furocoumarins/pharmacology , Photosensitizing Agents/pharmacology , Animals , Cell Culture Techniques , Cell Nucleus/drug effects , Cell Nucleus/radiation effects , Cells, Cultured , Female , HeLa Cells , Humans , Karyotyping , Male , Oocytes/cytology , Oocytes/drug effects , Oocytes/radiation effects , Spermatozoa/cytology , Spermatozoa/drug effects , Spermatozoa/radiation effects , Ultraviolet Rays , Xenopus laevis/embryology , Xenopus laevis/growth & development
17.
Proc Natl Acad Sci U S A ; 106(27): 11154-9, 2009 Jul 07.
Article in English | MEDLINE | ID: mdl-19549856

ABSTRACT

WT1, a critical regulator of kidney development, is a tumor suppressor for nephroblastoma but in some contexts functions as an oncogene. A limited number of direct transcriptional targets of WT1 have been identified to explain its complex roles in tumorigenesis and organogenesis. In this study we performed genome-wide screening for direct WT1 targets, using a combination of ChIP-ChIP and expression arrays. Promoter regions bound by WT1 were highly G-rich and resembled the sites for a number of other widely expressed transcription factors such as SP1, MAZ, and ZNF219. Genes directly regulated by WT1 were implicated in MAPK signaling, axon guidance, and Wnt pathways. Among directly bound and regulated genes by WT1, nine were identified in the Wnt signaling pathway, suggesting that WT1 modulates a subset of Wnt components and responsive genes by direct binding. To prove the biological importance of the interplay between WT1 and Wnt signaling, we showed that WT1 blocked the ability of Wnt8 to induce a secondary body axis during Xenopus embryonic development. WT1 inhibited TCF-mediated transcription activated by Wnt ligand, wild type and mutant, stabilized beta-catenin by preventing TCF4 loading onto a promoter. This was neither due to direct binding of WT1 to the TCF binding site nor to interaction between WT1 and TCF4, but by competition of WT1 and TCF4 for CBP. WT1 interference with Wnt signaling represents an important mode of its action relevant to the suppression of tumor growth and guidance of development.


Subject(s)
Genetic Testing , Genome/genetics , Signal Transduction/genetics , WT1 Proteins/metabolism , Wnt Proteins/metabolism , Animals , Base Sequence , Binding Sites , CREB-Binding Protein/metabolism , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA/metabolism , Embryo, Nonmammalian/metabolism , Embryonic Development , Gene Expression Profiling , Gene Expression Regulation , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic/genetics , Protein Binding , TCF Transcription Factors/metabolism , Transcription, Genetic , Xenopus/embryology
19.
Mol Carcinog ; 47(11): 835-44, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18404646

ABSTRACT

Pancreatic adeniocarcinoma is among the deadliest of human cancers. Apigenin, an antitumor flavonoid, inhibits pancreatic cancer cell proliferation in vitro. Geminin is a recently identified novel protein that plays a critical role in preventing abnormal DNA replication by binding to and inhibiting the essential replication factor Cdt1. Microarray analysis identified geminin to be downregulated in pancreatic cancer cells treated with apigenin. Therefore, we investigated the effects of apigenin on geminin expression and other proteins involved in replication (Cdc6, Cdt1, and MCM7) in pancreatic cancer cell lines CD18 and S2013. Real time RT-PCR and western blotting analysis showed that geminin expression is downregulated by apigenin at both mRNA and protein levels. Furthermore, treatment of cells with proteosome inhibitor MG132 reversed the downregulation of geminin by apigenin, supporting our hypothesis that the degradation pathway is another mechanism by which apigenin affects geminin expression. Apigenin treatment also resulted in downregulation of Cdc6 at both mRNA and protein levels. However, Cdt1 and MCM7 expression was not affected in apigenin-treated cells. The effect of apigenin treatment on geminin promoter activity was measured by transient transfection of Hela cells with a reporter gene, demonstrating that apigenin inhibited geminin promoter activity. Geminin expression was also evaluated in human pancreatic tissue (n = 15) by immunohistochemistry and showed that geminin is overexpressed in human pancreatic cancer compared to normal adjacent pancreatic tissue. In conclusion, our studies demonstrated that geminin is overexpressed in human pancreatic cancer and downregulated by apigenin which may contribute to the antitumor effect of this natural flavonoid.


Subject(s)
Apigenin/pharmacology , Biological Products/pharmacology , Cell Cycle Proteins/metabolism , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Pancreatic Neoplasms/metabolism , Cell Cycle Proteins/genetics , Cell Line, Tumor , Geminin , Humans , Leupeptins/pharmacology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Promoter Regions, Genetic/genetics , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors , RNA, Messenger/genetics
20.
Dent Today ; 27(12): 108, 110-1, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19133641

ABSTRACT

This patient's treatment involved a complex diagnostic challenge, as well as a challenging clinical sequence due to the utilization of immediate implant placement and restoration after extraction along with immediate prosthodontic restoration with sinus elevation with bone grafting. The inability to have diagnostic patient wax try-ins required a detailed and exacting diagnostic work-up which included significant laboratory diagnostic wax-ups. The utilization of the ACP Parameters of Care for Partial Edentulism for a PDI Class IV patient provided a framework in which care could be planned and executed with confidence. The availability of various reconstructive materials and techniques to create a seamless restorative result is essential to the success of this type of advanced treatment.


Subject(s)
Esthetics, Dental , Mouth Rehabilitation/methods , Alveolar Ridge Augmentation , Crowns , Dental Implants , Dental Prosthesis, Implant-Supported , Denture, Complete, Immediate , Denture, Complete, Lower , Humans , Male , Malocclusion/therapy , Mastication/physiology , Middle Aged , Patient Care Planning , Tooth Abrasion/therapy , Tooth Diseases/therapy , Tooth Eruption , Vertical Dimension
SELECTION OF CITATIONS
SEARCH DETAIL
...