Network


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

Hotspot


Dive into the research topics where Sargis Sedrakyan is active.

Publication


Featured researches published by Sargis Sedrakyan.


Stem Cells | 2008

Human Amniotic Fluid Stem Cells Can Integrate and Differentiate into Epithelial Lung Lineages

Gianni Carraro; Laura Perin; Sargis Sedrakyan; Stefano Giuliani; Caterina Tiozzo; Jooeun Lee; Gianluca Turcatel; Stijn De Langhe; Barbara Driscoll; Saverio Bellusci; Parviz Minoo; Anthony Atala; Roger E. De Filippo; David Warburton

A new source of stem cells has recently been isolated from amniotic fluid; these amniotic fluid stem cells have significant potential for regenerative medicine. These cells are multipotent, showing the ability to differentiate into cell types from each embryonic germ layer. We investigated the ability of human amniotic fluid stem cells (hAFSC) to integrate into murine lung and to differentiate into pulmonary lineages after injury. Using microinjection into cultured mouse embryonic lungs, hAFSC can integrate into the epithelium and express the early human differentiation marker thyroid transcription factor 1 (TTF1). In adult nude mice, following hyperoxia injury, tail vein‐injected hAFSC localized in the distal lung and expressed both TTF1 and the type II pneumocyte marker surfactant protein C. Specific damage of Clara cells through naphthalene injury produced integration and differentiation of hAFSC at the bronchioalveolar and bronchial positions with expression of the specific Clara cell 10‐kDa protein. These results illustrate the plasticity of hAFSC to respond in different ways to different types of lung damage by expressing specific alveolar versus bronchiolar epithelial cell lineage markers, depending on the type of injury to recipient lung.


Cell Proliferation | 2007

Renal differentiation of amniotic fluid stem cells

Laura Perin; Stefano Giuliani; D. Jin; Sargis Sedrakyan; Gianni Carraro; R. Habibian; David Warburton; Anthony Atala; R. E. De Filippo

Abstract.  Objectives: The role of stem cells in regenerative medicine is evolving rapidly. Here, we describe the application, for kidney regeneration, of a novel non‐genetically modified stem cell, derived from human amniotic fluid. We show that these pluripotent cells can develop and differentiate into de novo kidney structures during organogenesis in vitro. Materials and methods: Human amniotic fluid‐derived stem cells (hAFSCs) were isolated from human male amniotic fluid obtained between 12 and 18 weeks gestation. Green fluorescent protein and Lac‐Z‐transfected hAFSCs were microinjected into murine embryonic kidneys (12.5–18 days gestation) and were maintained in a special co‐culture system in vitro for 10 days. Techniques of live microscopy, histology, chromogenic in situ hybridization and reverse transcriptase polymerase chain reaction were used to characterize the hAFSCs during their integration and differentiation in concert with the growing organ. Results: Green fluorescent protein and Lac‐Z‐transfected hAFSCs demonstrated long‐term viability in organ culture. Histological analysis of injected kidneys revealed that hAFSCs were capable of contributing to the development of primordial kidney structures including renal vesicle, C‐ and S‐shaped bodies. Reverse transcriptase polymerase chain reaction confirmed expression of early kidney markers for: zona occludens‐1, glial‐derived neurotrophic factor and claudin. Conclusions: Human amniotic fluid‐derived stem cells may represent a potentially limitless source of ethically neutral, unmodified pluripotential cells for kidney regeneration.


Cell Metabolism | 2015

A Periodic Diet that Mimics Fasting Promotes Multi-System Regeneration, Enhanced Cognitive Performance, and Healthspan

Sebastian Brandhorst; In Young Choi; Min Wei; Chia Wei Cheng; Sargis Sedrakyan; Gerardo Navarrete; Louis Dubeau; Li Peng Yap; Ryan Park; Manlio Vinciguerra; Stefano Di Biase; Hamed Mirzaei; Mario G. Mirisola; Patra Childress; Lingyun Ji; Susan Groshen; Fabio Penna; Patrizio Odetti; Laura Perin; Peter S. Conti; Yuji Ikeno; Brian K. Kennedy; Pinchas Cohen; Todd E. Morgan; Tanya B. Dorff; Valter D. Longo

Prolonged fasting (PF) promotes stress resistance, but its effects on longevity are poorly understood. We show that alternating PF and nutrient-rich medium extended yeast lifespan independently of established pro-longevity genes. In mice, 4 days of a diet that mimics fasting (FMD), developed to minimize the burden of PF, decreased the size of multiple organs/systems, an effect followed upon re-feeding by an elevated number of progenitor and stem cells and regeneration. Bi-monthly FMD cycles started at middle age extended longevity, lowered visceral fat, reduced cancer incidence and skin lesions, rejuvenated the immune system, and retarded bone mineral density loss. In old mice, FMD cycles promoted hippocampal neurogenesis, lowered IGF-1 levels and PKA activity, elevated NeuroD1, and improved cognitive performance. In a pilot clinical trial, three FMD cycles decreased risk factors/biomarkers for aging, diabetes, cardiovascular disease, and cancer without major adverse effects, providing support for the use of FMDs to promote healthspan.


PLOS ONE | 2010

Protective effect of human amniotic fluid stem cells in an immunodeficient mouse model of acute tubular necrosis

Laura Perin; Sargis Sedrakyan; Stefano Giuliani; Stefano Da Sacco; Gianni Carraro; Liron Shiri; Kevin V. Lemley; Michael Rosol; Sam Wu; Anthony Atala; David Warburton; Roger E. De Filippo

Acute Tubular Necrosis (ATN) causes severe damage to the kidney epithelial tubular cells and is often associated with severe renal dysfunction. Stem-cell based therapies may provide alternative approaches to treating of ATN. We have previously shown that clonal c-kitpos stem cells, derived from human amniotic fluid (hAFSC) can be induced to a renal fate in an ex-vivo system. Herein, we show for the first time the successful therapeutic application of hAFSC in a mouse model with glycerol-induced rhabdomyolysis and ATN. When injected into the damaged kidney, luciferase-labeled hAFSC can be tracked using bioluminescence. Moreover, we show that hAFSC provide a protective effect, ameliorating ATN in the acute injury phase as reflected by decreased creatinine and BUN blood levels and by a decrease in the number of damaged tubules and apoptosis therein, as well as by promoting proliferation of tubular epithelial cells. We show significant immunomodulatory effects of hAFSC, over the course of ATN. We therefore speculate that AFSC could represent a novel source of stem cells that may function to modulate the kidney immune milieu in renal failure caused by ATN.


The Journal of Urology | 2010

Human Amniotic Fluid as a Potential New Source of Organ Specific Precursor Cells for Future Regenerative Medicine Applications

Stefano Da Sacco; Sargis Sedrakyan; Francesco Boldrin; Stefano Giuliani; PierPaolo Parnigotto; Rezvan Habibian; David Warburton; Roger E. De Filippo; Laura Perin

PURPOSE Human amniotic fluid contains multiple cell types, including pluripotent and committed progenitor cells, and fully differentiated cells. We characterized various cell populations in amniotic fluid. MATERIALS AND METHODS Optimum culture techniques for multiple cell line passages with minimal morphological change were established. Cell line analysis and characterization were done with reverse transcriptase and real-time polymerase chain reaction. Immunoseparation was done to distinguish native progenitor cell lines and their various subpopulations. RESULTS Endodermal and mesodermal marker expression was greatest in samples of early gestational age while ectodermal markers showed a constant rate across all samples. Pluripotent and mesenchymal cells were always present but hematopoietic cell markers were expressed only in older samples. Specific markers for lung, kidney, liver and heart progenitor cells were increasingly expressed after 18 weeks of gestation. We specifically focused on a CD24+OB-cadherin+ population that could identify uninduced metanephric mesenchyma-like cells, which in vivo are nephron precursors. The CD24+OB-cadherin+ cell line was isolated and subjected to further immunoseparation to select 5 distinct amniotic fluid kidney progenitor cell subpopulations based on E-cadherin, podocalyxin, nephrin, TRKA and PDGFRA expression, respectively. CONCLUSIONS These subpopulations may represent different precursor cell lineages committed to specific renal cell fates. Committed progenitor cells in amniotic fluid may provide an important and novel resource of useful cells for regenerative medicine purposes.


Methods in Cell Biology | 2008

Characterization of Human Amniotic Fluid Stem Cells and Their Pluripotential Capability

Laura Perin; Sargis Sedrakyan; Stafano Da Sacco; Roger E. De Filippo

Over the past decade, there has been ever-increasing emphasis placed on stem cells and their potential role in regenerative medicine for reconstruction of bio-artificial tissues and organs. Scientists have looked at various sources for pluripotential cells ranging from embryonic stem cells to adult stem cells. Amniocentesis is a well-established technique for the collection of cells derived from the human embryo. In this chapter, we are going to describe how to isolate, maintain in culture, and characterize the pluripotential capabilities of stem cells derived from amniocentesis in an in vitro and in vivo system. Cell samples are obtained from human pregnancies, and the progenitor cells are isolated from male fetuses with a normal karyotype in order to confirm the absence of maternal admixed cells. Progenitor cells express embryonic-specific cell markers, they show a high self-renewal capacity with 350 population doublings, and normal ploidy is confirmed by cell-cycle analyses. They maintain their undifferentiated state, pluripotential ability, clonogenicity, and telomere length over the population doublings. The progenitor cells are inducible to different cell lineages (osteogenic, adipogenic, skeletal muscle, endothelial, neuronal, and hepatic cells) under specific growth conditions. The ability to induce cell-type-specific differentiation is confirmed by phenotypic changes, immunocytochemistry, gene expression, and functional analyses. In addition, we will describe an application of these cells in an ex vivo and in vivo system for potential in organ (renal) regeneration. The progenitor cells described in this chapter have a high potential for expansion, and may be a good source for research and therapeutic applications where large numbers of cells are needed. Progenitor cells isolated during gestation may be beneficial for fetuses diagnosed with malformations and could be cryopreserved for future self-use.


Journal of The American Society of Nephrology | 2012

Injection of Amniotic Fluid Stem Cells Delays Progression of Renal Fibrosis

Sargis Sedrakyan; Stefano Da Sacco; Anna Milanesi; Liron Shiri; Astgik Petrosyan; Radka Varimezova; David Warburton; Kevin V. Lemley; Roger E. De Filippo; Laura Perin

Injection of amniotic fluid stem cells ameliorates the acute phase of acute tubular necrosis in animals by promoting proliferation of injured tubular cells and decreasing apoptosis, but whether these stem cells could be of benefit in CKD is unknown. Here, we used a mouse model of Alport syndrome, Col4a5(-/-) mice, to determine whether amniotic fluid stem cells could modify the course of progressive renal fibrosis. Intracardiac administration of amniotic fluid stem cells before the onset of proteinuria delayed interstitial fibrosis and progression of glomerular sclerosis, prolonged animal survival, and ameliorated the decline in kidney function. Treated animals exhibited decreased recruitment and activation of M1-type macrophages and a higher proportion of M2-type macrophages, which promote tissue remodeling. Amniotic fluid stem cells did not differentiate into podocyte-like cells and did not stimulate production of the collagen IVa5 needed for normal formation and function of the glomerular basement membrane. Instead, the mechanism of renal protection was probably the paracrine/endocrine modulation of both profibrotic cytokine expression and recruitment of macrophages to the interstitial space. Furthermore, injected mice retained a normal number of podocytes and had better integrity of the glomerular basement membrane compared with untreated Col4a5(-/-) mice. Inhibition of the renin-angiotensin system by amniotic fluid stem cells may contribute to these beneficial effects. In conclusion, treatment with amniotic fluid stem cells may be beneficial in kidney diseases characterized by progressive renal fibrosis.


Pediatric Research | 2008

Stem cell and regenerative science applications in the development of bioengineering of renal tissue.

Laura Perin; Stefano Giuliani; Sargis Sedrakyan; Stefano Da Sacco; Roger E. De Filippo

A rising number of patients with acute and chronic renal failure worldwide have created urgency for clinicians and investigators to search out alternative therapies other than chronic renal dialysis and/or organ transplantation. This review focuses on the recent achievements in this area, and discusses the various approaches in the development of bioengineering of renal tissue including recent discoveries in the field of regenerative medicine research and stem cells. A variety of stem cells, ranging from embryonic, bone marrow, endogenous, and amniotic fluid, have been investigated and may prove useful as novel alternatives for organ regeneration both in vitro and in vivo. Tissue engineering, developmental biology, and therapeutic cloning techniques have significantly contributed to our understanding of some of the molecular mechanisms involved in renal regeneration and have demonstrated that renal tissue can be generated de novo with similar physiologic functions as native tissue. Ultimately all of these emerging technologies may provide viable therapeutic options for regenerative medicine applications focused on the bioengineering of renal tissue for the future.


American Journal of Respiratory Cell and Molecular Biology | 2011

The Milieu of Damaged Alveolar Epithelial Type 2 Cells Stimulates Alveolar Wound Repair by Endogenous and Exogenous Progenitors

Sue Buckley; Wei Shi; Gianni Carraro; Sargis Sedrakyan; Stefano Da Sacco; Barbara Driscoll; Laura Perin; Roger E. De Filippo; David Warburton

Alveolar epithelial integrity is dependent upon the alveolar milieu, yet the milieu of the damaged alveolar epithelial cell type 2 (AEC2) has been little studied. Characterization of its components may offer the potential for ex vivo manipulation of stem cells to optimize their therapeutic potential. We examined the cytokine profile of AEC2 damage milieu, hypothesizing that it would promote endogenous epithelial repair while recruiting cells from other locations and instructing their engraftment and differentiation. Bronchoalveolar lavage and lung extract from hyperoxic rats represented AEC2 in vivo damage milieu, and medium from a scratch-damaged AEC2 monolayer represented in vitro damage. CINC-2 and ICAM, the major cytokines detected by proteomic cytokine array in AEC2 damage milieu, were chemoattractive to normoxic AECs and expedited in vitro wound healing, which was blocked by their respective neutralizing antibodies. The AEC2 damage milieu was also chemotactic for exogenous uncommitted human amniotic fluid stem cells (hAFSCs), increasing migration greater than 20-fold. hAFSCs attached within an in vitro AEC2 wound and expedited wound repair by contributing cytokines migration inhibitory factor and plasminogen activator inhibitor 1 to the AEC2 damage milieu, which promoted wound healing. The AEC2 damage milieu also promoted differentiation of a subpopulation of hAFSCs to express SPC, TTF-1, and ABCA3, phenotypic markers of distal alveolar epithelium. Thus, the microenvironment created by AEC2 damage not only promotes autocrine repair but also can attract uncommitted stem cells, which further augment healing through cytokine secretion and differentiation.


PLOS ONE | 2013

A Novel Source of Cultured Podocytes

Stefano Da Sacco; Kevin V. Lemley; Sargis Sedrakyan; Ilenia Zanusso; Astgik Petrosyan; Janos Peti-Peterdi; James L. Burford; Roger E. De Filippo; Laura Perin

Amniotic fluid is in continuity with multiple developing organ systems, including the kidney. Committed, but still stem-like cells from these organs may thus appear in amniotic fluid. We report having established for the first time a stem-like cell population derived from human amniotic fluid and possessing characteristics of podocyte precursors. Using a method of triple positive selection we obtained a population of cells (hAKPC-P) that can be propagated in vitro for many passages without immortalization or genetic manipulation. Under specific culture conditions, these cells can be differentiated to mature podocytes. In this work we compared these cells with conditionally immortalized podocytes, the current gold standard for in vitro studies. After in vitro differentiation, both cell lines have similar expression of the major podocyte proteins, such as nephrin and type IV collagen, that are characteristic of mature functional podocytes. In addition, differentiated hAKPC-P respond to angiotensin II and the podocyte toxin, puromycin aminonucleoside, in a way typical of podocytes. In contrast to immortalized cells, hAKPC-P have a more nearly normal cell cycle regulation and a pronounced developmental pattern of specific protein expression, suggesting their suitability for studies of podocyte development for the first time in vitro. These novel progenitor cells appear to have several distinct advantages for studies of podocyte cell biology and potentially for translational therapies.

Collaboration


Dive into the Sargis Sedrakyan's collaboration.

Top Co-Authors

Avatar

Laura Perin

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Roger E. De Filippo

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Stefano Da Sacco

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

David Warburton

Children's Hospital Los Angeles

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gianni Carraro

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Anthony Atala

Wake Forest Institute for Regenerative Medicine

View shared research outputs
Top Co-Authors

Avatar

Barbara Driscoll

Children's Hospital Los Angeles

View shared research outputs
Top Co-Authors

Avatar

Kevin V. Lemley

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Astgik Petrosyan

University of Southern California

View shared research outputs
Researchain Logo
Decentralizing Knowledge