Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Viral Immunol ; 37(3): 167-175, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38574259

RESUMO

Zika virus (ZIKV) is an emerging flavivirus associated with several neurological diseases such as Guillain-Barré syndrome in adults and microcephaly in newborn children. Its distribution and mode of transmission (via Aedes aegypti and Aedes albopictus mosquitoes) collectively cause ZIKV to be a serious concern for global health. High genetic homology of flaviviruses and shared ecology is a hurdle for accurate detection. Distinguishing infections caused by different viruses based on serological recognition can be misleading as many anti-flavivirus monoclonal antibodies (mAbs) discovered to date are highly cross-reactive, especially those against the envelope (E) protein. To provide more specific research tools, we produced ZIKV E directed hybridoma cell lines and characterized two highly ZIKV-specific mAb clones (mAbs A11 and A42) against several members of the Flavivirus genus. Epitope mapping of mAb A11 revealed glycan loop specificity in Domain I of the ZIKV E protein. The development of two highly specific mAbs targeting the surface fusion protein of ZIKV presents a significant advancement in research capabilities as these can be employed as essential tools to enhance our understanding of ZIKV identification on infected cells ex vivo or in culture.


Assuntos
Aedes , Flavivirus , Infecção por Zika virus , Zika virus , Animais , Recém-Nascido , Humanos , Proteínas do Envelope Viral , Anticorpos Monoclonais , Anticorpos Neutralizantes , Anticorpos Antivirais
2.
PLoS Pathog ; 19(5): e1011409, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37200377

RESUMO

The hallmark of severe COVID-19 involves systemic cytokine storm and multi-organ injury including testicular inflammation, reduced testosterone, and germ cell depletion. The ACE2 receptor is also expressed in the resident testicular cells, however, SARS-CoV-2 infection and mechanisms of testicular injury are not fully understood. The testicular injury could be initiated by direct virus infection or exposure to systemic inflammatory mediators or viral antigens. We characterized SARS-CoV-2 infection in different human testicular 2D and 3D culture systems including primary Sertoli cells, Leydig cells, mixed seminiferous tubule cells (STC), and 3D human testicular organoids (HTO). Data shows that SARS-CoV-2 does not productively infect any testicular cell type. However, exposure of STC and HTO to inflammatory supernatant from infected airway epithelial cells and COVID-19 plasma decreased cell viability and resulted in the death of undifferentiated spermatogonia. Further, exposure to only SARS-CoV-2 Envelope protein caused inflammatory response and cytopathic effects dependent on TLR2, while Spike 1 or Nucleocapsid proteins did not. A similar trend was observed in the K18-hACE2 transgenic mice which demonstrated a disrupted tissue architecture with no evidence of virus replication in the testis that correlated with peak lung inflammation. Virus antigens including Spike 1 and Envelope proteins were also detected in the serum during the acute stage of the disease. Collectively, these data strongly suggest that testicular injury associated with SARS-CoV-2 infection is likely an indirect effect of exposure to systemic inflammation and/or SARS-CoV-2 antigens. Data also provide novel insights into the mechanism of testicular injury and could explain the clinical manifestation of testicular symptoms associated with severe COVID-19.


Assuntos
COVID-19 , Masculino , Camundongos , Animais , Humanos , COVID-19/metabolismo , Testículo , SARS-CoV-2 , Efeito Espectador , Inflamação/metabolismo , Camundongos Transgênicos
3.
bioRxiv ; 2022 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-36172118

RESUMO

The hallmark of severe COVID-19 involves systemic cytokine storm and multi-organ failure including testicular injury and germ cell depletion. The ACE2 receptor is also expressed in the resident testicular cells however, SARS-CoV-2 infection and mechanisms of testicular injury are not fully understood. The testicular injury can likely result either from direct virus infection of resident cells or by exposure to systemic inflammatory mediators or virus antigens. We here characterized SARS-CoV-2 infection in different human testicular 2D and 3D models including primary Sertoli cells, Leydig cells, mixed seminiferous tubule cells (STC), and 3D human testicular organoids (HTO). Data shows that SARS-CoV-2 does not establish a productive infection in any testicular cell types. However, exposure of STC and HTO to inflammatory supernatant from infected airway epithelial cells and COVID-19 plasma depicted a significant decrease in cell viability and death of undifferentiated spermatogonia. Further, exposure to only SARS-CoV-2 envelope protein, but not Spike or nucleocapsid proteins led to cytopathic effects on testicular cells that was dependent on the TLR2 receptor. A similar trend was observed in the K18h-ACE2 mouse model which revealed gross pathology in the absence of virus replication in the testis. Collectively, data strongly indicates that the testicular injury is not due to direct infection of SARS-CoV-2 but more likely an indirect effect of exposure to systemic inflammation or SARS-CoV-2 antigens. Data also provide novel insights into the mechanism of testicular injury and could explain the clinical manifestation of testicular symptoms associated with severe COVID-19.

4.
Front Microbiol ; 13: 1062499, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36713156

RESUMO

Sexual transmission of Zika virus (ZIKV) is associated with virus persistence in the testes and shedding in the seminal fluid for months after recovery. We previously demonstrated that ZIKV can establish long-term replication without causing cytotoxicity in human Sertoli cells (SC), responsible for maintaining the immune privileged compartment of seminiferous tubules. Functional gene expression analyses also predicted activation of multiple virus sensing pathways including TLR3, RIG-I, and MDA5. Here, we elucidated which of the RNA virus sensing receptors play a decisive role in restricting ZIKV replication. We show that both poly I:C and IFN-ß treatment induced a robust antiviral state and reduced ZIKV replication significantly, suggesting that virus sensing and antiviral signaling are functional in SC. Silencing of TLR3, 7, and 9 did not affect virus replication kinetics; however, both RIG-I and MDA5 played a synergistic role in inducing an anti-ZIKV response. Further, the impact of SC-specific immunosuppressive pathways that collectively regulate SC function, specifically the TGF-ß superfamily members, TGF-ß, Activin A, and BMP6, on ZIKV replication was investigated. While ZIKV did not modulate the expression of TGF-ß and Activin A, BMP6 signaling was suppressed at later stages of infection. Notably, treatment with BMP6 increased IFN-ß, p-IRF3, and p-STAT1 levels, and expression of key interferon-stimulated genes including MDA5, suggesting that BMP6 enhances antiviral response in SC. Collectively, this study further delineates the key role of the RIG-I-like receptors in sensing ZIKV in SC, and reveals a novel role of BMP6 in modulating innate immune and antiviral response in the testes.

5.
Front Microbiol ; 12: 667146, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34079533

RESUMO

Zika virus (ZIKV) is unique among mosquito-borne flaviviruses in its ability to be sexually transmitted. The testes have been implicated as sites of long-term ZIKV replication, and our previous studies have identified Sertoli cells (SC), the nurse cells of the seminiferous epithelium that govern spermatogenesis, as major targets of ZIKV infection. To improve our understanding of the interaction of ZIKV with human SC, we analyzed ZIKV-induced proteome changes in these cells using high-throughput liquid chromatography-tandem mass spectrometry (LC-MS/MS). Our data demonstrated that interferon (IFN) signaling was the most significantly enriched pathway and the antiviral proteins MX1 and IFIT1 were among the top upregulated proteins in SC following ZIKV infection. The dynamic between IFN response and ZIKV infection kinetics in SC remains unclear, therefore we further determined whether MX1 and IFIT1 serve as antiviral effectors against ZIKV. We found that increased levels of MX1 at the later time points of infection coincided with diminished ZIKV infection while the silencing of MX1 and IFIT1 enhanced peak ZIKV propagation in SC. Furthermore, although IFN-I exposure was found to significantly hinder ZIKV replication in SC, IFN response was attenuated in these cells as compared to other cell types. The data in this study highlight IFN-I as a driver of the antiviral state that limits ZIKV infection in SC and suggests that MX1 and IFIT1 function as antiviral effectors against ZIKV in SC. Collectively, this study provides important biological insights into the response of SC to ZIKV infection and the ability of the virus to persist in the testes.

6.
mBio ; 10(4)2019 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-31311882

RESUMO

Zika virus (ZIKV) is unique among mosquito-borne flaviviruses in its ability to be sexually transmitted. Persistent ZIKV infection in the testes, which are immune privileged organs, long after peripheral clearance suggests involvement of immunosuppressive pathways; however, the underlying mechanisms remain undetermined. We recently demonstrated that ZIKV infects human Sertoli cells (SC), the major cell type of the seminiferous epithelium responsible for maintaining the immune privileged compartment of seminiferous tubules. Recent reports have identified the TAM (Tyro3, Axl, Mer) receptor tyrosine kinase Axl as an entry receptor and/or immune modulator for ZIKV in a cell type-specific manner. Interestingly, the seminiferous epithelium exhibits high basal expression of the Axl receptor where it is involved in clearance of apoptotic germ cells and immunosuppression. Here, we show that Axl was highly expressed in SC compared to Leydig cells (LC) that correlated with robust ZIKV infection of SC, but not LC. Further, neutralization of Axl receptor and its ligand Gas6 strongly attenuated virus entry in SC. However, inhibition of Axl kinase did not affect ZIKV entry but instead led to decreased protein levels of suppressor of cytokine signaling 1 (SOCS1) and SOCS3, increased expression of interferon-stimulated genes (ISGs), and reduced ZIKV replication. Similarly, treatment of multicellular human testicular organoids with an Axl kinase inhibitor attenuated ZIKV replication and increased ISG expression. Together, our data demonstrate that Axl promotes ZIKV entry and negatively regulates the antiviral state of SC to augment ZIKV infection of the testes and provides new insights into testis antiviral immunity and ZIKV persistence.IMPORTANCE Recent Zika virus (ZIKV) outbreaks have identified sexual transmission as a new route of disease spread not reported for other flaviviruses. ZIKV crosses the blood-testis barrier and establishes infection in seminiferous tubules, the site for spermatozoa development. Currently, there are no therapies to treat ZIKV infection, and the immune mechanisms underlying testicular persistence are unclear. We found that multiple human testicular cell types, except Leydig cells, support ZIKV infection. Axl receptor, which plays a pivotal role in maintaining the immunosuppressive milieu of the testis, is highly expressed in Sertoli cells and augments ZIKV infection by promoting virus entry and negatively regulating the antiviral state. By using testicular organoids, we further describe the antiviral role of Axl inhibition. The significance of our research lies in defining cross talk between Axl and type I interferon signaling as an essential mechanism of immune control that can inform therapeutic efforts to clear ZIKV from the testis.


Assuntos
Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Células de Sertoli/metabolismo , Células de Sertoli/virologia , Internalização do Vírus , Infecção por Zika virus/metabolismo , Infecção por Zika virus/virologia , Zika virus/fisiologia , Células Cultivadas , Resistência à Doença , Interações Hospedeiro-Patógeno , Humanos , Masculino , Infecção por Zika virus/imunologia , Receptor Tirosina Quinase Axl
7.
Sci Rep ; 8(1): 8702, 2018 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-29880853

RESUMO

Confirmed reports of Zika virus (ZIKV) in seminal fluid months after clearance of viremia suggests that ZIKV can establish persistent infection in the seminiferous tubules, an immune privileged site of the testis. The seminiferous tubule epithelium is mainly composed of Sertoli cells that function to nourish and protect developing germ cells. We recently demonstrated that primary human Sertoli cells (hSeC) were highly susceptible to ZIKV as compared to dengue virus without causing cell death and thus may act as a reservoir for ZIKV in the testes. However, the cellular and immune responses of hSeC to infection with ZIKV or any other virus are not yet characterized. Using genome-wide RNA-seq to compare immunoprofiles of hSeC, we show that the most prominent response to ZIKV at early stage of infection was suppression of cell growth and proliferation functional pathways. Peak virus replication was associated with induction of multiple antiviral defense pathways. Unique ZIKV-associated signatures included dysregulation of germ cell-Sertoli cell junction signaling. This study demonstrates that hSeC are capable of signaling through canonical pro-inflammatory pathways and provides insights into unique cell-type-specific response induced by ZIKV in association with viral persistence in the testes.


Assuntos
Interações Hospedeiro-Patógeno/imunologia , Células de Sertoli/imunologia , Infecção por Zika virus/imunologia , Zika virus/fisiologia , Linhagem Celular , Humanos , Masculino , Células de Sertoli/patologia , Células de Sertoli/virologia , Infecção por Zika virus/patologia
9.
J Virol ; 91(22)2017 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-28878076

RESUMO

Confirmed reports of Zika virus (ZIKV) in human seminal fluid for months after the clearance of viremia suggest the ability of ZIKV to establish persistent infection in the seminiferous tubules, an immune-privileged site in the testis protected by the blood-testis barrier, also called the Sertoli cell (SC) barrier (SCB). However, cellular targets of ZIKV in human testis and mechanisms by which the virus enters seminiferous tubules remain unclear. We demonstrate that primary human SCs were highly susceptible to ZIKV compared to the closely related dengue virus and induced the expression of alpha interferon (IFN-α), key cytokines, and cell adhesion molecules (vascular cell adhesion molecule 1 [VCAM-1] and intracellular adhesion molecule 1 [ICAM-1]). Furthermore, using an in vitro SCB model, we show that ZIKV was released on the adluminal side of the SCB model with a higher efficiency than in the blood-brain barrier model. ZIKV-infected SCs exhibited enhanced adhesion of leukocytes that correlated with decreases in SCB integrity. ZIKV infection did not affect the expression of tight and adherens junction proteins such as ZO-1, claudin, and JAM-A; however, exposure of SCs to inflammatory mediators derived from ZIKV-infected macrophages led to the degradation of the ZO-1 protein, which correlated with increased SCB permeability. Taken together, our data suggest that infection of SCs may be one of the crucial steps by which ZIKV gains access to the site of spermatozoon development and identify SCs as a therapeutic target to clear testicular infections. The SCB model opens up opportunities to assess interactions of SCs with other testicular cells and to test the ability of anti-ZIKV drugs to cross the barrier.IMPORTANCE Recent outbreaks of ZIKV, a neglected mosquito-borne flavivirus, have identified sexual transmission as a new route of disease spread, which has not been reported for other flaviviruses. To be able to sexually transmit for months after the clearance of viremia, ZIKV must establish infection in the seminiferous tubules, the site of spermatozoon development. However, little is known about the cell types that support ZIKV infection in the human testis. Currently, there are no models to study mechanisms of virus persistence in the seminiferous tubules. We provide evidence that ZIKV infection of human Sertoli cells, which are an important component of the seminiferous tubules, is robust and induces a strong antiviral response. The use of an in vitro Sertoli cell barrier to describe how ZIKV or inflammatory mediators derived from ZIKV-infected macrophages compromise barrier integrity will enable studies to explore the interactions of other testicular cells with Sertoli cells and to test novel antivirals for clearing testicular ZIKV infection.


Assuntos
Barreira Hematotesticular/imunologia , Células de Sertoli/imunologia , Infecção por Zika virus/imunologia , Zika virus/imunologia , Barreira Hematotesticular/patologia , Barreira Hematotesticular/virologia , Moléculas de Adesão Celular/imunologia , Células Cultivadas , Claudinas/imunologia , Dengue/imunologia , Dengue/patologia , Vírus da Dengue/imunologia , Humanos , Interferon-alfa/imunologia , Macrófagos/imunologia , Macrófagos/patologia , Masculino , Receptores de Superfície Celular/imunologia , Células de Sertoli/patologia , Células de Sertoli/virologia , Molécula 1 de Adesão de Célula Vascular/imunologia , Infecção por Zika virus/patologia , Proteína da Zônula de Oclusão-1/imunologia
10.
Front Microbiol ; 8: 1571, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28861075

RESUMO

Ebola virus (EBOV), a member of the Filoviridae family, causes the most severe form of viral hemorrhagic fever. Although no FDA licensed vaccine or treatment against Ebola virus disease (EVD) is currently available, Ebola virus glycoprotein (GP) is the major antigen used in all candidate Ebola vaccines. Recent reports of protection as quickly as within 6 days of administration of the rVSV-based vaccine expressing EBOV GP before robust humoral responses were generated suggests that the innate immune responses elicited early after vaccination may contribute to the protection. However, the innate immune responses induced by EBOV GP in the absence of viral vectors or adjuvants have not been fully characterized in vivo. Our recent studies demonstrated that immunization with highly purified recombinant GP in the absence of adjuvants induced a robust IgG response and partial protection against EBOV infection suggesting that GP alone can induce protective immunity. In this study we investigated the early immune response to purified EBOV GP alone in vitro and in vivo. We show that GP was efficiently internalized by antigen presenting cells and subsequently induced production of key inflammatory cytokines. In vivo, immunization of mice with EBOV GP triggered the production of key Th1 and Th2 innate immune cytokines and chemokines, which directly governed the recruitment of CD11b+ macrophages and CD11c+ dendritic cells to the draining lymph nodes (DLNs). Pre-treatment of mice with a TLR4 antagonist inhibited GP-induced cytokine production and recruitment of immune cells to the DLN. EBOV GP also upregulated the expression of costimulatory molecules in bone marrow derived macrophages suggesting its ability to enhance APC stimulatory capacity, which is critical for the induction of effective antigen-specific adaptive immunity. Collectively, these results provide the first in vivo evidence that early innate immune responses to EBOV GP are mediated via the TLR4 pathway and are able to modulate the innate-adaptive interface. These mechanistic insights into the adjuvant-like property of EBOV GP may help to develop a better understanding of how optimal prophylactic efficacy of EBOV vaccines can be achieved as well as further explore the potential post-exposure use of vaccines to prevent filoviral disease.

11.
J Mater Sci Mater Med ; 25(3): 681-90, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24408274

RESUMO

Mechanically robust and biomimicking scaffolds are needed for structural engineering of tissues such as the intervertebral disc, which are prone to failure and incapable of natural healing. Here, the formation of thick, randomly aligned polycaprolactone electrospun fibre structures infiltrated with alginate is reported. The composites are characterised using both indentation and tensile testing and demonstrate substantially different tensile and compressive moduli. The composites are mechanically robust and exhibit large strains-to-failure, exhibiting toughening mechanisms observed in other composite material systems. The method presented here provides a way to create large-scale biomimetic scaffolds that more closely mimic the composite structure of natural tissue, with tuneable tensile and compressive properties via the fibre and matrix phases, respectively.


Assuntos
Alginatos/química , Materiais Biomiméticos/síntese química , Eletroquímica/métodos , Hidrogéis/química , Poliésteres/química , Força Compressiva , Módulo de Elasticidade , Ácido Glucurônico/química , Dureza , Ácidos Hexurônicos/química , Teste de Materiais , Rotação , Estresse Mecânico , Resistência à Tração
12.
J Mech Behav Biomed Mater ; 11: 16-26, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22658151

RESUMO

Tissue engineering offers a paradigm shift in the treatment of back pain. Engineered intervertebral discs could replace degenerated tissue and overcome the limitations of current treatments, which substantially alter the biomechanical properties of the spine. The centre of the disc, the nucleus pulposus, is an amorphous gel with a large bound water content and it can resist substantial compressive loads. Due to similarities in their compositions, hydrogels have frequently been considered as substitutes for the nucleus pulposus. However, there has been limited work characterising the time-dependent mechanical behaviour of hydrogel scaffolds for nucleus pulposus tissue engineering. Poroelastic behaviour, which plays a key role in nutrient transport, is of particular importance. Here, we investigate the time-dependent mechanical properties of gelatin and agar hydrogels and of gelatin-agar composites. The time-dependent properties of these hydrogels are explored using viscoelastic and poroelastic frameworks. Several gel formulations demonstrate comparable equilibrium elastic behaviour to the nucleus pulposus under unconfined compression, but permeability values that are much greater than those of the native tissue. A range of time-dependent responses are observed in the composite gels examined, presenting the opportunity for targeted design of custom hydrogels with combinations of mechanical properties optimized for tissue engineering applications.


Assuntos
Hidrogéis/química , Disco Intervertebral/citologia , Fenômenos Mecânicos , Engenharia Tecidual/métodos , Ágar/química , Força Compressiva , Elasticidade , Gelatina/química , Teste de Materiais , Fatores de Tempo , Viscosidade
13.
Nat Mater ; 11(7): 642-9, 2012 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-22635042

RESUMO

To investigate how substrate properties influence stem-cell fate, we cultured single human epidermal stem cells on polydimethylsiloxane (PDMS) and polyacrylamide (PAAm) hydrogel surfaces, 0.1 kPa-2.3 MPa in stiffness, with a covalently attached collagen coating. Cell spreading and differentiation were unaffected by polydimethylsiloxane stiffness. However, cells on polyacrylamide of low elastic modulus (0.5 kPa) could not form stable focal adhesions and differentiated as a result of decreased activation of the extracellular-signal-related kinase (ERK)/mitogen-activated protein kinase (MAPK) signalling pathway. The differentiation of human mesenchymal stem cells was also unaffected by PDMS stiffness but regulated by the elastic modulus of PAAm. Dextran penetration measurements indicated that polyacrylamide substrates of low elastic modulus were more porous than stiff substrates, suggesting that the collagen anchoring points would be further apart. We then changed collagen crosslink concentration and used hydrogel-nanoparticle substrates to vary anchoring distance at constant substrate stiffness. Lower collagen anchoring density resulted in increased differentiation. We conclude that stem cells exert a mechanical force on collagen fibres and gauge the feedback to make cell-fate decisions.


Assuntos
Técnicas de Cultura de Células/métodos , Diferenciação Celular , Matriz Extracelular/metabolismo , Células-Tronco Mesenquimais/citologia , Resinas Acrílicas/química , Resinas Acrílicas/farmacologia , Diferenciação Celular/efeitos dos fármacos , Colágeno/química , Dimetilpolisiloxanos/química , Dimetilpolisiloxanos/farmacologia , Matriz Extracelular/efeitos dos fármacos , Humanos , Recém-Nascido , Queratinócitos/citologia , Fenômenos Mecânicos , Células-Tronco Mesenquimais/efeitos dos fármacos
14.
Acta Biomater ; 7(10): 3586-94, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21723962

RESUMO

Hydroxyapatite-gelatin composites have been proposed as suitable scaffolds for bone and dentin tissue regeneration. There is considerable interest in producing these scaffolds using biomimetic methods due to their low energy costs and potential to create composites similar to the tissues they are intended to replace. Here an existing process used to coat a surface with hydroxyapatite under near physiological conditions, the alternate soaking process, is modified and automated using an inexpensive "off the shelf" robotics kit. The process is initially used to precipitate calcium phosphate coatings. Then, in contrast to previous utilizations of the alternate soaking process, gelatin was added directly to the solutions in order to co-precipitate hydroxyapatite-gelatin composites. Samples were investigated by Fourier transform infrared spectroscopy, scanning electron microscopy, energy dispersive X-ray spectroscopy and nanoindentation. Calcium phosphate coatings formed by the alternate soaking process exhibited different calcium to phosphate ratios, with correspondingly distinct structural morphologies. The coatings demonstrated an interconnected structure with measurable mechanical properties, even though they were 95% porous. In contrast, hydroxyapatite-gelatin composite coatings over 2mm thick could be formed with little visible porosity. The hydroxyapatite-gelatin composites demonstrate a composition and mechanical properties similar to those of cortical bone.


Assuntos
Materiais Biomiméticos/farmacologia , Biomimética/métodos , Osso e Ossos/efeitos dos fármacos , Tecidos Suporte/química , Automação , Durapatita/química , Gelatina/farmacologia , Microscopia Eletrônica de Varredura , Soluções , Espectroscopia de Infravermelho com Transformada de Fourier , Temperatura
15.
Spine J ; 10(7): 602-9, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20547110

RESUMO

BACKGROUND CONTEXT: Discectomy is a common procedure for treating sciatica. However, both the operation and preceding herniated disc alter the biomechanical properties of the spinal segment. The disc mechanics are also altered in patients with chronic contained herniation. The biomechanical properties of the disc can potentially be restored with an elastomeric nucleus replacement implanted via minimally invasive surgery. PURPOSE: The purpose of this study was to determine whether the compressive characteristics of the intervertebral disc after a nucleotomy can be restored with an elastomeric nucleus replacement. STUDY DESIGN: A finite element model of the L4-L5 intervertebral disc was created to investigate the effect of the implantation of an elastomeric nucleus replacement on the biomechanical properties of the disc under axial loading. METHOD: A L4-L5 physiologic intervertebral disc model was constructed and then modified to contain a range by volume of nucleotomies and nucleus replacements. The material properties of the nucleus replacement were based on experimental data for an elastomeric implant. The compressive stiffness, radial annular bulge, and stress distribution of the nucleotomy and nucleus replacement models were investigated under displacement-controlled loading. RESULTS: Removal of nucleus pulposus from the physiologic disc reduced the force necessary to compress the disc 2 mm by 50%, altered the von Mises stress distribution, and reduced the outward radial annular bulge. Replacing the natural nucleus pulposus of the physiologic disc with an artificial nucleus reduced the force required to compress the disc 2 mm by 10%, indicating a restoration of disc compressive stiffness. The von Mises stress distribution and annular bulge observed in the disc with an artificial nucleus were similar to that observed in the physiologic disc. CONCLUSION: This study demonstrates that despite having different material properties, a nucleus replacement implant can restore the axial compressive mechanical properties of a disc after a discectomy. The implant carries compressive load and transfers the load into annular hoop stress.


Assuntos
Artroplastia de Substituição/instrumentação , Discotomia/métodos , Análise de Elementos Finitos , Deslocamento do Disco Intervertebral/cirurgia , Disco Intervertebral/cirurgia , Vértebras Lombares/cirurgia , Força Compressiva , Simulação por Computador , Humanos , Disco Intervertebral/fisiopatologia , Deslocamento do Disco Intervertebral/fisiopatologia , Teste de Materiais , Desenho de Prótese , Falha de Prótese , Recuperação de Função Fisiológica , Estresse Mecânico , Suporte de Carga/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...