Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Indian J Ophthalmol ; 70(11): 4057-4059, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36308158

RESUMO

Cataract surgery is constantly evolving with new techniques, machines, and procedures entering the lexicon. With the modernization, the cost of surgery is increasing with respect to the surgeon, patient, and the society. Early rehabilitation with modern minimal access techniques reduces the societal cost of intervention. Manual small-incision surgery is simple, safer, and cheaper when contrasted with the cost and steep learning curve of machine-driven surgeries. A nucleus of a normal size is 6 mm, which can be bisected; 3 mm longitudinal fragments can be removed through 3.5 to 4.5 mm incision, and a large nucleus of 9.0 mm can trisected and removed through 3.5 to 4.5 mm. The limbal tunnel incision is 3.5 mm, which is close to 2.8 mm phacoincision, so it gives all the advantages of modern phacosurgery. The search of an ideal technique for manual phaco-fragmentation leads us to this specially designed snare (designed by the first author AS) which can tackle any grade nucleus. The nucleus can be bisected or trisected with ease. The technique has been around for 2 decades. There is a small learning curve. The complications are few and can be minimized with practice and simulation. It is a safe, valid, repeatable, and generalizable surgical procedure.


Assuntos
Extração de Catarata , Catarata , Oftalmologia , Humanos , Extração de Catarata/métodos , Microcirurgia/métodos , Curva de Aprendizado
2.
Indian J Ophthalmol ; 70(8): 3008-3014, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35918962

RESUMO

Purpose: The present study compares the efficacy, safety, and immunogenicity of Lupin's biosimilar ranibizumab with that of Lucentis® in patients with neovascular age-related macular degeneration. Methods: This prospective, double-blind, multi-centric phase-III study was conducted across 19 centers in India. A total of 202 patients with neovascular age-related macular degeneration were randomized (1:1) to receive either Lupin's biosimilar ranibizumab or Lucentis®, 0.5 mg, as an intravitreous injection once every month for 3 months. The primary efficacy endpoint was the proportion of patients who lost fewer than 15 letters from baseline in best-corrected visual acuity. The safety profile included assessment of adverse events, ophthalmic examination, physical and systemic examination, and vital parameters. The immunogenicity assessment was based on evaluation of anti-drug antibodies. Results: Overall, 174 patients (87 [86.14%] in each group) completed the study. The demographics and baseline characteristics were comparable between the treatment groups. The proportion of patients losing fewer than 15 letters from baseline best corrected visual acuity score in the study eye was comparable between two groups. The difference between Lupin's ranibizumab and Lucentis® for the proportion of patients who lost fewer than 15 letters was within the predefined equivalence margin (intention-to-treat population: 1.0%; 95% confidence interval [CI], -3.3% to 5.4% and per protocol population: 1.2%; 95% CI, -3.2% to 6.4%). The incidence of treatment-emergent adverse events was comparable, and 11 (10.89%) patients in Lupin's ranibizumab and 19 (18.81%) patients in Lucentis® group had at least one treatment-emergent adverse event. The immunogenicity incidence as assessed by proportion of patients with positive anti-drug antibodies was numerically lower in Lupin's ranibizumab (4.95%) than Lucentis® (12.87%). Conclusion: Lupin's biosimilar ranibizumab demonstrated therapeutic equivalence, desirable safety, and favorable immunogenicity profile compared to Lucentis®.


Assuntos
Medicamentos Biossimilares , Degeneração Macular , Degeneração Macular Exsudativa , Inibidores da Angiogênese , Anticorpos Monoclonais Humanizados , Humanos , Injeções Intravítreas , Degeneração Macular/induzido quimicamente , Estudos Prospectivos , Ranibizumab , Resultado do Tratamento , Acuidade Visual , Degeneração Macular Exsudativa/diagnóstico , Degeneração Macular Exsudativa/tratamento farmacológico
3.
Am Heart J ; 185: 26-34, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28267472

RESUMO

The relative outcomes of intravascular ultrasound (IVUS)-guided percutaneous coronary intervention (PCI) compared with angiography-guided PCI with drug-eluting stent (DES) in complex lesions have not been established. We sought to compare the efficacy and safety of IVUS-guided PCI with angiography-guided PCI in patients with complex coronary lesions treated with DES. METHODS: Electronic databases were searched to identify all randomized trials comparing IVUS-guided vs angiography-guided DES implantation. We evaluated major adverse cardiac events (MACE), all-cause and cardiovascular death, myocardial infarction, target lesion revascularization (TLR), target vessel revascularization (TVR), and stent thrombosis outcomes at the longest reported follow-up. Random-effects modeling was used to calculate pooled relative risk (RR) and 95% CIs. RESULTS: Eight trials comprising 3,276 patients (1,635 IVUS-guided and 1,641 angiography-guided) enrolling only patients with complex lesions were included. Mean follow-up was 1.4±0.5years. Compared with angiography-guided PCI, patients undergoing IVUS-guided PCI had significantly lower MACE (RR 0.64, 95% CI 0.51-0.80, P=.0001), TLR (RR 0.62, 95% CI 0.45-0.86, P=.004), and TVR (RR 0.60, 95% CI 0.42-0.87, P=.007). There were no significant differences for stent thrombosis, cardiovascular death, or all-cause death. In meta-regression analysis, IVUS-guided PCI was of greatest benefit in reducing MACE in patients with acute coronary syndromes, diabetes, and long lesions. CONCLUSIONS: The present meta-analysis demonstrates a significant reduction in MACE, TVR, and TLR with IVUS-guided DES implantation in complex coronary lesions.


Assuntos
Angiografia Coronária , Doença da Artéria Coronariana/cirurgia , Stents Farmacológicos , Intervenção Coronária Percutânea/métodos , Cirurgia Assistida por Computador/métodos , Ultrassonografia de Intervenção , Doenças Cardiovasculares/mortalidade , Causas de Morte , Doença da Artéria Coronariana/diagnóstico por imagem , Oclusão de Enxerto Vascular/epidemiologia , Humanos , Mortalidade , Infarto do Miocárdio/epidemiologia , Revascularização Miocárdica/estatística & dados numéricos , Ensaios Clínicos Controlados Aleatórios como Assunto , Trombose/epidemiologia
4.
PLoS One ; 10(11): e0142337, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26560447

RESUMO

Hepatocellular carcinoma (HCC) is the most common form of liver cancer (~80%), and it is one of the few cancer types with rising incidence in the United States. This highly invasive cancer is very difficult to detect until its later stages, resulting in limited treatment options and low survival rates. There is a dearth of knowledge regarding the mechanisms associated with the effects of biomechanical forces such as interstitial fluid flow (IFF) on hepatocellular carcinoma invasion. We hypothesized that interstitial fluid flow enhanced hepatocellular carcinoma cell invasion through chemokine-mediated autologous chemotaxis. Utilizing a 3D in vitro invasion assay, we demonstrated that interstitial fluid flow promoted invasion of hepatocellular carcinoma derived cell lines. Furthermore, we showed that autologous chemotaxis influences this interstitial fluid flow-induced invasion of hepatocellular carcinoma derived cell lines via the C-X-C chemokine receptor type 4 (CXCR4)/C-X-C motif chemokine 12 (CXCL12) signaling axis. We also demonstrated that mitogen-activated protein kinase (MEK)/extracellular signal-regulated kinase (ERK) signaling affects interstitial fluid flow-induced invasion; however, this pathway was separate from CXCR4/CXCL12 signaling. This study demonstrates, for the first time, the potential role of interstitial fluid flow in hepatocellular carcinoma invasion. Uncovering the mechanisms that control hepatocellular carcinoma invasion will aid in enhancing current liver cancer therapies and provide better treatment options for patients.


Assuntos
Carcinoma Hepatocelular/metabolismo , Quimiocina CXCL12/metabolismo , Líquido Extracelular/metabolismo , Neoplasias Hepáticas/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Invasividade Neoplásica/patologia , Receptores CXCR4/metabolismo , Transdução de Sinais/fisiologia , Animais , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Quimiotaxia/fisiologia , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Neoplasias Hepáticas/patologia , Masculino , Fosforilação , Ratos , Ratos Sprague-Dawley
5.
J Neurochem ; 135(6): 1113-22, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26465636

RESUMO

Breast cancer resistance protein (Bcrp/Abcg2) localized at the blood-brain barrier (BBB) limits permeability into the brain of many xenobiotics, including pharmacological agents. Peroxisome proliferator-activated receptor α (Pparα), a ligand-activated transcription factor, primarily involved in lipid metabolism, has been shown to regulate the functional expression of Bcrp in human cerebral microvascular endothelial cells (hCMEC/D3). The aim of this study was to investigate ex vivo and in vivo, the regulation of Bcrp by Pparα in an intact BBB. Ex vivo quantitative real-time PCR and immunoblot analyses showed significant up-regulation of Abcg2/Bcrp mRNA and protein levels in CD-1 mouse brain capillaries incubated with clofibrate, a Pparα ligand. Fluorescence-based transport assays in CD-1 and C57BL/6 brain capillaries showed that exposure to clofibrate significantly increased Bcrp transport activity. This increase was not observed in capillaries isolated from Pparα knockout mice. In vivo, we found: i) significant Bcrp protein up-regulation in clofibrate-dosed CD-1 and C57BL/6 capillary lysates, but no effect in Pparα knockout capillary lysates, and ii) significantly increased Bcrp transport activity in capillaries isolated from clofibrate-treated mice. These results demonstrate an increase in Bcrp functional expression by Pparα in brain capillaries, and suggest that Pparα is another nuclear receptor that can contribute to the regulation of membrane efflux transporters and drug permeability at the BBB. We propose the involvement of the following pathways in clofibrate-mediated induction of the drug transporter Abcg2/Bcrp mRNA, protein expression and function by the nuclear receptor Pparα, in mouse brain capillary endothelial cells. Upon activation with clofibrate (Pparα, ligand), Pparα complex translocates from the cytoplasm into the nucleus and further recruits coactivators and transcription machinery which induce the transcription of Abcg2 gene and ultimately results in upregulation of Bcrp protein expression and function. These findings have significant implications since Bcrp is known to play an important role at the BBB in preventing the permeability of several xenobiotics and drugs into the brain.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Neoplasias da Mama/metabolismo , PPAR alfa/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Transporte Biológico/fisiologia , Neoplasias da Mama/genética , Capilares/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica/fisiologia , Proteínas de Membrana Transportadoras/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Citoplasmáticos e Nucleares/metabolismo
6.
Exp Hematol ; 43(12): 1047-61, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26391449

RESUMO

Forward genetic screens in zebrafish have been used to identify genes essential for the generation of primitive blood and the emergence of hematopoietic stem cells (HSCs), but have not elucidated the genes essential for hematopoietic stem and progenitor cell (HSPC) proliferation and differentiation because of the lack of methodologies to functionally assess these processes. We previously described techniques used to test the developmental potential of HSPCs by culturing them on zebrafish kidney stromal (ZKS) cells, derived from the main site of hematopoiesis in the adult teleost. Here we describe an additional primary stromal cell line we refer to as zebrafish embryonic stromal trunk (ZEST) cells, derived from tissue surrounding the embryonic dorsal aorta, the site of HSC emergence in developing fish. ZEST cells encouraged HSPC differentiation toward the myeloid, lymphoid, and erythroid pathways when assessed by morphologic and quantitative reverse transcription polymerase chain reaction analyses. Additionally, ZEST cells significantly expanded the number of cultured HSPCs in vitro, indicating that these stromal cells are supportive of both HSPC proliferation and multilineage differentiation. Examination of ZEST cells indicates that they express numerous cytokines and Notch ligands and possess endothelial characteristics. Further characterization of ZEST cells should prove to be invaluable in understanding the complex signaling cascades instigated by the embryonic hematopoietic niche required to expand and differentiate HSPCs. Elucidating these processes and identifying possibilities for the modulation of these molecular pathways should allow the in vitro expansion of HSPCs for a multitude of therapeutic uses.


Assuntos
Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Rim/metabolismo , Células Estromais/metabolismo , Peixe-Zebra/metabolismo , Animais , Sobrevivência Celular/fisiologia , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Células-Tronco Hematopoéticas/citologia , Rim/citologia , Células Estromais/citologia
7.
J Vis Exp ; (65)2012 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-22872144

RESUMO

The growth and progression of most solid tumors depend on the initial transformation of the cancer cells and their response to stroma-associated signaling in the tumor microenvironment (1). Previously, research on the tumor microenvironment has focused primarily on tumor-stromal interactions (1-2). However, the tumor microenvironment also includes a variety of biophysical forces, whose effects remain poorly understood. These forces are biomechanical consequences of tumor growth that lead to changes in gene expression, cell division, differentiation and invasion(3). Matrix density (4), stiffness (5-6), and structure (6-7), interstitial fluid pressure (8), and interstitial fluid flow (8) are all altered during cancer progression. Interstitial fluid flow in particular is higher in tumors compared to normal tissues (8-10). The estimated interstitial fluid flow velocities were measured and found to be in the range of 0.1-3 µm s(-1), depending on tumor size and differentiation (9, 11). This is due to elevated interstitial fluid pressure caused by tumor-induced angiogenesis and increased vascular permeability (12). Interstitial fluid flow has been shown to increase invasion of cancer cells (13-14), vascular fibroblasts and smooth muscle cells (15). This invasion may be due to autologous chemotactic gradients created around cells in 3-D (16) or increased matrix metalloproteinase (MMP) expression (15), chemokine secretion and cell adhesion molecule expression (17). However, the mechanism by which cells sense fluid flow is not well understood. In addition to altering tumor cell behavior, interstitial fluid flow modulates the activity of other cells in the tumor microenvironment. It is associated with (a) driving differentiation of fibroblasts into tumor-promoting myofibroblasts (18), (b) transporting of antigens and other soluble factors to lymph nodes (19), and (c) modulating lymphatic endothelial cell morphogenesis (20). The technique presented here imposes interstitial fluid flow on cells in vitro and quantifies its effects on invasion (Figure 1). This method has been published in multiple studies to measure the effects of fluid flow on stromal and cancer cell invasion (13-15, 17). By changing the matrix composition, cell type, and cell concentration, this method can be applied to other diseases and physiological systems to study the effects of interstitial flow on cellular processes such as invasion, differentiation, proliferation, and gene expression.


Assuntos
Técnicas de Cultura de Células/métodos , Líquido Extracelular/metabolismo , Melanoma/metabolismo , Melanoma/patologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Corantes Fluorescentes/química , Humanos , Indóis/química , Microscopia de Fluorescência/métodos , Invasividade Neoplásica , Metástase Neoplásica , Microambiente Tumoral
8.
J Assoc Physicians India ; 58: 198-9, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20848824

RESUMO

We present a case of a left sided pheochromocytoma, who had normal levels of 24 urinary vanilylmandelic acid and a normal MIBG scan. The diagnosis was confirmed on histopathology of the adrenal gland.


Assuntos
Neoplasias das Glândulas Suprarrenais/patologia , Feocromocitoma/patologia , 3-Iodobenzilguanidina , Neoplasias das Glândulas Suprarrenais/diagnóstico por imagem , Neoplasias das Glândulas Suprarrenais/cirurgia , Adrenalectomia , Adulto , Reações Falso-Negativas , Feminino , Humanos , Feocromocitoma/diagnóstico por imagem , Feocromocitoma/cirurgia , Cintilografia , Compostos Radiofarmacêuticos , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA