Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Arvydas Maminishkis is active.

Publication


Featured researches published by Arvydas Maminishkis.


The FASEB Journal | 2010

MicroRNA-204/211 alters epithelial physiology

F. Wang; Connie Zhang; Arvydas Maminishkis; Lijin Dong; Connie Zhi; Rong Li; Jing Zhao; Vladimir Majerciak; Arti B. Gaur; Shan Chen; Sheldon S. Miller

MicroRNA (miRNA) expression in fetal human retinal pigment epithelium (hfRPE), retina, and choroid were pairwise compared to determine those miRNAs that are enriched by 10‐fold or more in each tissue compared with both of its neighbors. miRs‐184, 187, 200a/200b, 204/211, and 221/222 are enriched in hfRPE by 10‐ to 754‐fold compared with neuroretina or choroid (P<0.05). Five of these miRNAs are enriched in RPE compared with 20 tissues throughout the body and are 10‐ to 20,000‐fold more highly expressed (P<0.005). miR‐204 and 211 are the most highly expressed in the RPE. In addition, expression of miR‐204/211 is significantly lower in the NCI60 tumor cell line panel compared with that in 13 normal tissues, suggesting the progressive disruption of epithelial barriers and increased proliferation. We demonstrated that TGF‐β receptor 2 (TGF‐βR2) and SNAIL2 are direct targets of miR‐204 and that a reduction in miR‐204 expression leads to reduced expression of claudins 10, 16, and 19 (message/protein) consistent with our observation that anti‐miR‐204/211 decreased transepithelial resistance by 80% and reduced cell membrane voltage and conductance. The anti‐miR‐204‐induced decrease in Kir7.1 protein levels suggests a signaling pathway that connects TGF‐βR2 and maintenance of potassium homeostasis. Overall, these data indicate a critical role for miR‐204/211 in maintaining epithelial barrier function and cell physiology.—Wang, F. E., Zhang, C., Maminishkis, A., Dong, L., Zhi, C., Li, R., Zhao, J., Majerciak, V., Gaur, A. B., Chen, S., Miller, S. S. MicroRNA‐204/211 alters epithelial physiology. FASEB J. 24, 1552–1571 (2010). www.fasebj.org


Human Molecular Genetics | 2010

Transcriptome analysis and molecular signature of human retinal pigment epithelium

N.V. Strunnikova; Arvydas Maminishkis; J.J. Barb; F. Wang; C. Zhi; Yuri V. Sergeev; Wei-Min Chen; Albert O. Edwards; Dwight Stambolian; Gonçalo R. Abecasis; Anand Swaroop; Peter J. Munson; S.S. Miller

Retinal pigment epithelium (RPE) is a polarized cell layer critical for photoreceptor function and survival. The unique physiology and relationship to the photoreceptors make the RPE a critical determinant of human vision. Therefore, we performed a global expression profiling of native and cultured human fetal and adult RPE and determined a set of highly expressed ‘signature’ genes by comparing the observed RPE gene profiles to the Novartis expression database (SymAtlas: http://wombat.gnf.org/index.html) of 78 tissues. Using stringent selection criteria of at least 10-fold higher expression in three distinct preparations, we identified 154 RPE signature genes, which were validated by qRT-PCR analysis in RPE and in an independent set of 11 tissues. Several of the highly expressed signature genes encode proteins involved in visual cycle, melanogenesis and cell adhesion and Gene ontology analysis enabled the assignment of RPE signature genes to epithelial channels and transporters (ClCN4, BEST1, SLCA20) or matrix remodeling (TIMP3, COL8A2). Fifteen RPE signature genes were associated with known ophthalmic diseases, and 25 others were mapped to regions of disease loci. An evaluation of the RPE signature genes in a recently completed AMD genomewide association (GWA) data set revealed that TIMP3, GRAMD3, PITPNA and CHRNA3 signature genes may have potential roles in AMD pathogenesis and deserve further examination. We propose that RPE signature genes are excellent candidates for retinal diseases and for physiological investigations (e.g. dopachrome tautomerase in melanogenesis). The RPE signature gene set should allow the validation of RPE-like cells derived from human embryonic or induced pluripotent stem cells for cell-based therapies of degenerative retinal diseases.


Investigative Ophthalmology & Visual Science | 2008

Control of Chemokine Gradients by the Retinal Pigment Epithelium

Guangpu Shi; Arvydas Maminishkis; T. Banzon; S. Jalickee; Rong Li; Jeffrey A. Hammer; Sheldon S. Miller

PURPOSE Proinflammatory cytokines in degenerative diseases can lead to the loss of normal physiology and the destruction of surrounding tissues. In the present study, the physiological responses of human fetal retinal pigment epithelia (hfRPE) were examined in vitro after polarized activation of proinflammatory cytokine receptors. METHODS Primary cultures of hfRPE were stimulated with an inflammatory cytokine mixture (ICM): interleukin (IL)-1beta, tumor necrosis factor (TNF)-alpha, and interferon (IFN)-gamma. Western blot analysis and immunofluorescence were used to determine the expression/localization of the cytokine receptors on hfRPE. Polarized secretion of cytokines was measured. A capacitance probe technique was used to measure transepithelial fluid flow (J(V)) and resistance (R(T)). RESULTS IL-1R1 was mainly localized to the apical membrane and TNFR1 to the basal membrane, whereas IFN-gammaR1 was detected on both membranes. Activation by apical ICM induced a significant secretion of angiogenic and angiostatic chemokines, mainly across the hfRPE apical membrane. Addition of the ICM to the basal but not the apical bath significantly increased net fluid absorption (J(V)) across the hfRPE within 20 minutes. Similar increases in J(V) were produced by a 24-hour exposure to ICM, which significantly decreased total R(T). CONCLUSIONS Chemokine gradients across the RPE can be altered (1) through an ICM-induced change in polarized chemokine secretion and (2) through an increase in ICM-induced net fluid absorption. In vivo, both of these factors could contribute to the development of chemokine gradients that help mediate the progression of inflammation/angiogenesis at the retina/RPE/choroid complex.


Science Translational Medicine | 2014

IL-18 Attenuates Experimental Choroidal Neovascularization as a Potential Therapy for Wet Age-Related Macular Degeneration

Sarah L. Doyle; Ema Ozaki; Kiva Brennan; Marian M. Humphries; Kelly Mulfaul; James Keaney; Paul F. Kenna; Arvydas Maminishkis; Anna-Sophia Kiang; Sean P. Saunders; Emily Hams; Ed C. Lavelle; Clair M. Gardiner; Padraic G. Fallon; Peter Adamson; Peter Humphries; Matthew Campbell

IL-18 prevents choroidal neovascularization, the hallmark pathology of wet AMD, and is not toxic to the retinal pigment epithelium. Treating Age-Related Macular Degeneration with IL-18 Age-related macular degeneration (AMD) comes in two forms: “wet” and “dry.” Dry AMD is characterized by the death of various eye cells, whereas wet AMD arises from the formation of new blood vessels in the choroid. The only treatment for AMD is antibodies against growth factors that regulate vascular growth; this is not a cure, but rather a chronic therapy requiring direct injection into the eye. Seeking out a new therapy, Doyle and colleagues found that an inflammatory cytokine, interleukin-18 (IL-18), works to prevent neovascularization in animal models. The eyes of mice were hit with a laser to induce blood vessel growth, or choroidal neovascularization (CNV), which mimics the human AMD pathology. Recombinant, mature IL-18 injected into the eyes of mice or subcutaneously limited CNV formation. It has been suggested that IL-18 is toxic, but Doyle et al. showed that this is not the case through in vitro and in vivo studies; the “pro” form of the cytokine is, however, and can be regulated by autophagy. Because IL-18 is being tested in patients with cancer, the authors believe that this could translate and readily complement existing antiangiogenic strategies currently used for AMD, such as the antibody-based therapies targeting vascular endothelial growth factor (VEGF). Age-related macular degeneration (AMD) is the most common form of central retinal blindness globally. Distinct processes of the innate immune system, specifically activation of the NLRP3 inflammasome, have been shown to play a central role in the development of both “dry” and neovascular (“wet”) forms of the disease. We show that the inflammatory cytokine interleukin-18 (IL-18) can regulate choroidal neovascularization formation in mice. We observed that exogenous administration of mature recombinant IL-18 has no effect on retinal pigment epithelial (RPE) cell viability, but that overexpression of pro–IL-18 or pro–IL-1β alone can cause RPE cell swelling and subsequent atrophy, a process that can be inhibited by the promotion of autophagy. A direct comparison of local and systemic administration of mature recombinant IL-18 with current anti-VEGF (vascular endothelial growth factor)–based therapeutic strategies shows that IL-18 treatment works effectively alone and more effectively in combination with anti-VEGF therapy and represents a novel therapeutic strategy for the treatment of wet AMD.


Journal of Immunology | 2008

Gene Expression Profiling in Autoimmune Noninfectious Uveitis Disease

Zhuqing Li; Baoying Liu; Arvydas Maminishkis; Sankaranarayana P. Mahesh; Yeh S; Lew J; Lim Wk; Sen Hn; Clarke G; Ronald R. Buggage; Miller Ss; Robert B. Nussenblatt

Noninfectious uveitis is a predominantly T cell-mediated autoimmune, intraocular inflammatory disease. To characterize the gene expression profile from patients with noninfectious uveitis, PBMCs were isolated from 50 patients with clinically characterized noninfectious uveitis syndrome. A pathway-specific cDNA microarray was used for gene expression profiling and real-time PCR array for further confirmation. Sixty-seven inflammation- and autoimmune-associated genes were found differentially expressed in uveitis patients, with 28 of those genes being validated by real-time PCR. Several genes previously unknown for autoimmune uveitis, including IL-22, IL-19, IL-20, and IL-25/IL-17E, were found to be highly expressed among uveitis patients compared with the normal subjects with IL-22 expression highly variable among the patients. Furthermore, we show that IL-22 can affect primary human retinal pigment epithelial cells by decreasing total tissue resistance and inducing apoptosis possibly by decreasing phospho-Bad level. In addition, the microarray data identified a possible uveitis-associated gene expression pattern, showed distinct gene expression profiles in patients during periods of clinical activity and quiescence, and demonstrated similar expression patterns in related patients with similar clinical phenotypes. Our data provide the first evidence that a subset of IL-10 family genes are implicated in noninfectious uveitis and that IL-22 can affect human retinal pigment epithelial cells. The results may facilitate further understanding of the molecular mechanisms of autoimmune uveitis and other autoimmune originated inflammatory diseases.


Proceedings of the National Academy of Sciences of the United States of America | 2010

PDGF-CC blockade inhibits pathological angiogenesis by acting on multiple cellular and molecular targets

Xu Hou; Anil Kumar; Chunsik Lee; Bin Wang; Pachiappan Arjunan; Lijin Dong; Arvydas Maminishkis; Zhongshu Tang; Yang Li; Fan Zhang; Shi-Zhuang Zhang; Piotr Wardega; Sagarika Chakrabarty; Baoying Liu; Zhijian Wu; Peter Colosi; Robert N. Fariss; Johan Lennartsson; Robert B. Nussenblatt; J. Silvio Gutkind; Yihai Cao; Xuri Li

The importance of identifying VEGF-independent pathways in pathological angiogenesis is increasingly recognized as a result of the emerging drug resistance to anti-VEGF therapies. PDGF-CC is the third member of the PDGF family discovered after more than two decades of studies on PDGF-AA and PDGF-BB. The biological function of PDGF-CC and the underlying cellular and molecular mechanisms remain largely unexplored. Here, using different animal models, we report that PDGF-CC inhibition by neutralizing antibody, shRNA, or genetic deletion suppressed both choroidal and retinal neovascularization. Importantly, we revealed that PDGF-CC targeting acted not only on multiple cell types important for pathological angiogenesis, such as vascular mural and endothelial cells, macrophages, choroidal fibroblasts and retinal pigment epithelial cells, but also on the expression of other important angiogenic genes, such as PDGF-BB and PDGF receptors. At a molecular level, we found that PDGF-CC regulated glycogen synthase kinase (GSK)–3β phosphorylation and expression both in vitro and in vivo. Activation of GSK3β impaired PDGF-CC–induced angiogenesis, and inhibition of GSK3β abolished the antiangiogenic effect of PDGF-CC blockade. Thus, we identified PDGF-CC as an important candidate target gene for antiangiogenic therapy, and PDGF-CC inhibition may be of therapeutic value in treating neovascular diseases.


Journal of Biological Chemistry | 2012

Microphthalmia-associated Transcription Factor (MITF) Promotes Differentiation of Human Retinal Pigment Epithelium (RPE) by Regulating microRNAs-204/211 Expression

Jeffrey Adijanto; John J. Castorino; Zi-Xuan Wang; Arvydas Maminishkis; Gerald B. Grunwald; Nancy J. Philp

Background: microRNAs 204/211 regulate retinal pigment epithelial cell phenotype. Results: In RPE, MITF regulates miR-204/211 expression and down-regulation of MITF results in loss of RPE phenotype, which can be prevented by overexpressing miR-204/211. Conclusion: MITF-mediated expression of miR-204/211 directs RPE differentiation. Significance: miR-204/211-based therapeutics may be effective treatments for diseases that involve loss of RPE phenotype. The retinal pigment epithelium (RPE) plays a fundamental role in maintaining visual function and dedifferentiation of RPE contributes to the pathophysiology of several ocular diseases. To identify microRNAs (miRNAs) that may be involved in RPE differentiation, we compared the miRNA expression profiles of differentiated primary human fetal RPE (hfRPE) cells to dedifferentiated hfRPE cells. We found that miR-204/211, the two most highly expressed miRNAs in the RPE, were significantly down-regulated in dedifferentiated hfRPE cells. Importantly, transfection of pre-miR-204/211 into hfRPE cells promoted differentiation whereas adding miR-204/211 inhibitors led to their dedifferentiation. Microphthalmia-associated transcription factor (MITF) is a key regulator of RPE differentiation that was also down-regulated in dedifferentiated hfRPE cells. MITF knockdown decreased miR-204/211 expression and caused hfRPE dedifferentiation. Significantly, co-transfection of MITF siRNA with pre-miR-204/211 rescued RPE phenotype. Collectively, our data show that miR-204/211 promote RPE differentiation, suggesting that miR-204/211-based therapeutics may be effective treatments for diseases that involve RPE dedifferentiation such as proliferative vitreoretinopathy.


Journal of Immunology | 2011

Autoreactive Memory CD4+ T Lymphocytes That Mediate Chronic Uveitis Reside in the Bone Marrow through STAT3-Dependent Mechanisms

Hyun-Mee Oh; Cheng-Rong Yu; Y. Lee; Chi-Chao Chan; Arvydas Maminishkis; Charles E. Egwuagu

Organ-specific autoimmune diseases are usually characterized by repeated cycles of remission and recurrent inflammation. However, where the autoreactive memory T cells reside in between episodes of recurrent inflammation is largely unknown. In this study, we have established a mouse model of chronic uveitis characterized by progressive photoreceptor cell loss, retinal degeneration, focal retinitis, retinal vasculitis, multifocal choroiditis, and choroidal neovascularization, providing for the first time to our knowledge a useful model for studying long-term pathological consequences of chronic inflammation of the neuroretina. We show that several months after inception of acute uveitis, autoreactive memory T cells specific to retinal autoantigen, interphotoreceptor retinoid-binding protein (IRBP), relocated to bone marrow (BM). The IRBP-specific memory T cells (IL-7RαHighLy6CHighCD4+) resided in BM in resting state but upon restimulation converted to IL-17/IFN-γ–expressing effectors (IL-7RαLowLy6CLowCD4+) that mediated uveitis. We further show that T cells from STAT3-deficient (CD4-STAT3KO) mice are defective in α4β1 and osteopontin expression, defects that correlated with inability of IRBP-specific memory CD4-STAT3KO T cells to traffic into BM. We adoptively transferred uveitis to naive mice using BM cells from wild-type mice with chronic uveitis but not BM cells from CD4-STAT3KO, providing direct evidence that memory T cells that mediate uveitis reside in BM and that STAT3-dependent mechanism may be required for migration into and retention of memory T cells in BM. Identifying BM as a survival niche for T cells that cause uveitis suggests that BM stromal cells that provide survival signals to autoreactive memory T cells and STAT3-dependent mechanisms that mediate their relocation into BM are attractive therapeutic targets that can be exploited to selectively deplete memory T cells that drive chronic inflammation.


Investigative Ophthalmology & Visual Science | 2009

Expression, localization, and function of junctional adhesion molecule-C (JAM-C) in human retinal pigment epithelium.

Matina Economopoulou; Jeffrey A. Hammer; Fei Wang; Robert N. Fariss; Arvydas Maminishkis; Sheldon S. Miller

PURPOSE To determine the localization of JAM-C in human RPE and characterize its functions. METHODS Immunofluorescence, Western blot, and PCR was used to identify the localization and expression of JAM-C, ZO-1, N-cadherin, and ezrin in cultures of human fetal RPE (hfRPE) with or without si-RNA mediated JAM-C knockdown and in adult native RPE wholemounts. A transepithelial migration assay was used to study the migration of leukocytes through the hfRPE monolayer. RESULTS JAM-C localized at the tight junctions of cultured hfRPE cells and adult native RPE. During initial junction formation JAM-C was recruited to the primordial cell-cell contacts and after JAM-C knockdown, the organization of N-cadherin and ZO-1 at those contacts was disrupted. JAM-C knockdown caused a delay in the hfRPE cell polarization, as shown by reduced apical staining of ezrin. JAM-C inhibition significantly decreased the chemokine-induced transmigration of granulocytes but not monocytes through the hfRPE monolayer. CONCLUSIONS JAM-C localizes specifically in the tight junctions of hfRPE and adult native RPE. It is important for tight junction formation in hfRPE, possibly by regulating the recruitment of N-cadherin and ZO-1 at the cell-cell contacts, and has a role in the polarization of hfRPE cells. Finally, JAM-C promotes the basal-to-apical transmigration of granulocytes but not monocytes through the hfRPE monolayer.


Stem Cells Translational Medicine | 2014

A Multiplex High-Throughput Gene Expression Assay to Simultaneously Detect Disease and Functional Markers in Induced Pluripotent Stem Cell-Derived Retinal Pigment Epithelium

Marc Ferrer; Barbara Corneo; Janine Davis; Qin Wan; Kiyoharu Miyagishima; Rebecca King; Arvydas Maminishkis; Juan J. Marugan; Ruchi Sharma; Michael Shure; Sally Temple; Sheldon S. Miller; Kapil Bharti

There is continuing interest in the development of lineage‐specific cells from induced pluripotent stem (iPS) cells for use in cell therapies and drug discovery. Although in most cases differentiated cells show features of the desired lineage, they retain fetal gene expression and do not fully mature into “adult‐like” cells. Such cells may not serve as an effective therapy because, once implanted, immature cells pose the risk of uncontrolled growth. Therefore, there is a need to optimize lineage‐specific stem cell differentiation protocols to produce cells that no longer express fetal genes and have attained “adult‐like” phenotypes. Toward that goal, it is critical to develop assays that simultaneously measure cell function and disease markers in high‐throughput format. Here, we use a multiplex high‐throughput gene expression assay that simultaneously detects endogenous expression of multiple developmental, functional, and disease markers in iPS cell‐derived retinal pigment epithelium (RPE). We optimized protocols to differentiate iPS cell‐derived RPE that was then grown in 96‐ and 384‐well plates. As a proof of principle, we demonstrate differential expression of eight genes in iPS cells, iPS cell‐derived RPE at two different differentiation stages, and primary human RPE using this multiplex assay. The data obtained from the multiplex gene expression assay are significantly correlated with standard quantitative reverse transcription‐polymerase chain reaction‐based measurements, confirming the ability of this high‐throughput assay to measure relevant gene expression changes. This assay provides the basis to screen for compounds that improve RPE function and maturation and target disease pathways, thus providing the basis for effective treatments of several retinal degenerative diseases.

Collaboration


Dive into the Arvydas Maminishkis's collaboration.

Top Co-Authors

Avatar

Sheldon S. Miller

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Kapil Bharti

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Rong Li

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

T. Banzon

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Qin Wan

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Vladimir Khristov

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

F. Wang

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

C. Zhi

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Kiyoharu Miyagishima

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Balendu Shekhar Jha

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge