Network


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

Hotspot


Dive into the research topics where Alan Nguyen is active.

Publication


Featured researches published by Alan Nguyen.


Cellular and Molecular Life Sciences | 2014

Natural history of mesenchymal stem cells, from vessel walls to culture vessels

Iain R. Murray; Christopher C. West; Winters R. Hardy; Aaron W. James; Tea Soon Park; Alan Nguyen; Tulyapruek Tawonsawatruk; Lorenza Lazzari; Chia Soo; Bruno Péault

Abstract Mesenchymal stem/stromal cells (MSCs) can regenerate tissues by direct differentiation or indirectly by stimulating angiogenesis, limiting inflammation, and recruiting tissue-specific progenitor cells. MSCs emerge and multiply in long-term cultures of total cells from the bone marrow or multiple other organs. Such a derivation in vitro is simple and convenient, hence popular, but has long precluded understanding of the native identity, tissue distribution, frequency, and natural role of MSCs, which have been defined and validated exclusively in terms of surface marker expression and developmental potential in culture into bone, cartilage, and fat. Such simple, widely accepted criteria uniformly typify MSCs, even though some differences in potential exist, depending on tissue sources. Combined immunohistochemistry, flow cytometry, and cell culture have allowed tracking the artifactual cultured mesenchymal stem/stromal cells back to perivascular anatomical regions. Presently, both pericytes enveloping microvessels and adventitial cells surrounding larger arteries and veins have been described as possible MSC forerunners. While such a vascular association would explain why MSCs have been isolated from virtually all tissues tested, the origin of the MSCs grown from umbilical cord blood remains unknown. In fact, most aspects of the biology of perivascular MSCs are still obscure, from the emergence of these cells in the embryo to the molecular control of their activity in adult tissues. Such dark areas have not compromised intents to use these cells in clinical settings though, in which purified perivascular cells already exhibit decisive advantages over conventional MSCs, including purity, thorough characterization and, principally, total independence from in vitro culture. A growing body of experimental data is currently paving the way to the medical usage of autologous sorted perivascular cells for indications in which MSCs have been previously contemplated or actually used, such as bone regeneration and cardiovascular tissue repair.


Stem Cells and Development | 2012

Brief Review of Models of Ectopic Bone Formation

Michelle A. Scott; Benjamin Levi; Asal Askarinam; Alan Nguyen; Todd Rackohn; Kang Ting; Chia Soo; Aaron W. James

Ectopic bone formation is a unique biologic entity--distinct from other areas of skeletal biology. Animal research models of ectopic bone formation most often employ rodent models and have unique advantages over orthotopic (bone) environments, including a relative lack of bone cytokine stimulation and cell-to-cell interaction with endogenous (host) bone-forming cells. This allows for relatively controlled in vivo experimental bone formation. A wide variety of ectopic locations have been used for experimentation, including subcutaneous, intramuscular, and kidney capsule transplantation. The method, benefits and detractions of each method are summarized in the following review. Briefly, subcutaneous implantation is the simplest method. However, the most pertinent concern is the relative paucity of bone formation in comparison to other models. Intramuscular implantation is also widely used and relatively simple, however intramuscular implants are exposed to skeletal muscle satellite progenitor cells. Thus, distinguishing host from donor osteogenesis becomes challenging without cell-tracking studies. The kidney capsule (perirenal or renal capsule) method is less widely used and more technically challenging. It allows for supraphysiologic blood and nutrient resource, promoting robust bone growth. In summary, ectopic bone models are extremely useful in the evaluation of bone-forming stem cells, new osteoinductive biomaterials, and growth factors; an appropriate choice of model, however, will greatly increase experimental success.


Stem Cells Translational Medicine | 2012

An Abundant Perivascular Source of Stem Cells for Bone Tissue Engineering

Aaron W. James; Janette N. Zara; Mirko Corselli; Asal Askarinam; Ann M. Zhou; Alireza Hourfar; Alan Nguyen; Silva Megerdichian; Greg Asatrian; Shen Pang; David A. Stoker; Xinli Zhang; Benjamin M. Wu; Kang Ting; Bruno Péault; Chia Soo

Adipose tissue is an ideal mesenchymal stem cell (MSC) source, as it is dispensable and accessible with minimal morbidity. However, the stromal vascular fraction (SVF) of adipose tissue is a heterogeneous cell population, which has disadvantages for tissue regeneration. In the present study, we prospectively purified human perivascular stem cells (PSCs) from n = 60 samples of human lipoaspirate and documented their frequency, viability, and variation with patient demographics. PSCs are a fluorescence‐activated cell sorting‐sorted population composed of pericytes (CD45−, CD146+, CD34−) and adventitial cells (CD45−, CD146−, CD34+), each of which we have previously reported to have properties of MSCs. Here, we found that PSCs make up, on average, 43.2% of SVF from human lipoaspirate (19.5% pericytes and 23.8% adventitial cells). These numbers were minimally changed by age, gender, or body mass index of the patient or by length of refrigerated storage time between liposuction and processing. In a previous publication, we observed that human PSCs (hPSCs) formed significantly more bone in vivo in comparison with unsorted human SVF (hSVF) in an intramuscular implantation model. We now extend this finding to a bone injury model, observing that purified hPSCs led to significantly greater healing of mouse critical‐size calvarial defects than hSVF (60.9% healing as opposed to 15.4% healing at 2 weeks postoperative by microcomputed tomography analysis). These studies suggest that adipose‐derived hPSCs are a new cell source for future efforts in skeletal regenerative medicine. Moreover, hPSCs are a stem cell‐based therapeutic that is readily approvable by the U.S. Food and Drug Administration, with potentially increased safety, purity, identity, potency, and efficacy.


Frontiers in Physiology | 2013

A review of hedgehog signaling in cranial bone development

Angel Pan; Le Chang; Alan Nguyen; Aaron W. James

During craniofacial development, the Hedgehog (HH) signaling pathway is essential for mesodermal tissue patterning and differentiation. The HH family consists of three protein ligands: Sonic Hedgehog (SHH), Indian Hedgehog (IHH), and Desert Hedgehog (DHH), of which two are expressed in the craniofacial complex (IHH and SHH). Dysregulations in HH signaling are well documented to result in a wide range of craniofacial abnormalities, including holoprosencephaly (HPE), hypotelorism, and cleft lip/palate. Furthermore, mutations in HH effectors, co-receptors, and ciliary proteins result in skeletal and craniofacial deformities. Cranial suture morphogenesis is a delicate developmental process that requires control of cell commitment, proliferation and differentiation. This review focuses on both what is known and what remains unknown regarding HH signaling in cranial suture morphogenesis and intramembranous ossification. As demonstrated from murine studies, expression of both SHH and IHH is critical to the formation and fusion of the cranial sutures and calvarial ossification. SHH expression has been observed in the cranial suture mesenchyme and its precise function is not fully defined, although some postulate SHH to delay cranial suture fusion. IHH expression is mainly found on the osteogenic fronts of the calvarial bones, and functions to induce cell proliferation and differentiation. Unfortunately, neonatal lethality of IHH deficient mice precludes a detailed examination of their postnatal calvarial phenotype. In summary, a number of basic questions are yet to be answered regarding domains of expression, developmental role, and functional overlap of HH morphogens in the calvaria. Nevertheless, SHH and IHH ligands are integral to cranial suture development and regulation of calvarial ossification. When HH signaling goes awry, the resultant suite of morphologic abnormalities highlights the important roles of HH signaling in cranial development.


International Journal of Surgical Pathology | 2015

Clinical and Histopathological Diagnosis of Glomus Tumor: An Institutional Experience of 138 Cases

Marco Mravic; Gregory Y. LaChaud; Alan Nguyen; Michelle A. Scott; Sarah M. Dry; Aaron W. James

Background. Glomus tumors are relatively uncommon subcentimeteric benign perivascular neoplasms usually located on the fingers. With their blue-red color and common subungual location, they are commonly confused for vascular or melanocytic lesions. To date there is no comprehensive review of an institutional experience with glomus tumors. Methods. A 14-year retrospective review of all cases within University of California, Los Angeles, with either a clinical or pathological diagnosis of glomus tumor was performed. Data obtained included demographic information, tumor description, pathological diagnoses, immunohistochemical studies, radiographic and treatment information, and clinical course. Rates of concordance between clinical and pathological diagnoses and an evaluation of overlap with other entities were assessed. Results. Clinical diagnosis of glomus tumor showed concordance with a histopathological diagnosis (45.4% of cases). The most common alternate clinical diagnoses included lipoma, cyst, or angioma. A pathological diagnosis of glomus tumor was most common in the fourth to seventh decades of life. The most common presentation was a subcentimeter lesion on the digit. Deep-seated tumors had a strikingly increased risk for malignancy (33%). Radiological studies were not relied on frequently (18.2% of cases). Immunohistochemical analysis showed diffuse αSMA and MSA expression in nearly all cases (99% and 95%, respectively), with focal to diffuse CD34 immunostaining in 32% of cases. Discussion. Our study illustrates trends in the clinical versus pathologic diagnoses of glomus tumor, common competing diagnoses, a difference in demographics than is commonly reported (older age groups most commonly affected), and important differences in the use adjunctive diagnostic tools including radiology and immunohistochemistry.


Stem Cells Translational Medicine | 2014

Human Perivascular Stem Cell-Based Bone Graft Substitute Induces Rat Spinal Fusion

Choon G. Chung; Aaron W. James; Greg Asatrian; Le Chang; Alan Nguyen; Khoi Le; Georgina Bayani; Robert Lee; David A. Stoker; Shen Pang; Xinli Zhang; Kang Ting; Bruno Péault; Chia Soo

Adipose tissue is an attractive source of mesenchymal stem cells (MSCs) because of its abundance and accessibility. We have previously defined a population of native MSCs termed perivascular stem cells (PSCs), purified from diverse human tissues, including adipose tissue. Human PSCs (hPSCs) are a bipartite cell population composed of pericytes (CD146+CD34−CD45−) and adventitial cells (CD146−CD34+CD45−), isolated by fluorescence‐activated cell sorting and with properties identical to those of culture identified MSCs. Our previous studies showed that hPSCs exhibit improved bone formation compared with a sample‐matched unpurified population (termed stromal vascular fraction); however, it is not known whether hPSCs would be efficacious in a spinal fusion model. To investigate, we evaluated the osteogenic potential of freshly sorted hPSCs without culture expansion and differentiation in a rat model of posterolateral lumbar spinal fusion. We compared increasing dosages of implanted hPSCs to assess for dose‐dependent efficacy. All hPSC treatment groups induced successful spinal fusion, assessed by manual palpation and microcomputed tomography. Computerized biomechanical simulation (finite element analysis) further demonstrated bone fusion with hPSC treatment. Histological analyses showed robust endochondral ossification in hPSC‐treated samples. Finally, we confirmed that implanted hPSCs indeed differentiated into osteoblasts and osteocytes; however, the majority of the new bone formation was of host origin. These results suggest that implanted hPSCs positively regulate bone formation via direct and paracrine mechanisms. In summary, hPSCs are a readily available MSC population that effectively forms bone without requirements for culture or predifferentiation. Thus, hPSC‐based products show promise for future efforts in clinical bone regeneration and repair.


American Journal of Pathology | 2016

Novel Wnt Regulator NEL-Like Molecule-1 Antagonizes Adipogenesis and Augments Osteogenesis Induced by Bone Morphogenetic Protein 2

Jia Shen; Aaron W. James; Xinli Zhang; Shen Pang; Janette N. Zara; Greg Asatrian; Michael Chiang; Min Lee; Kevork Khadarian; Alan Nguyen; Kevin S. Lee; Ronald K. Siu; Sotirios Tetradis; Kang Ting; Chia Soo

The differentiation factor NEL-like molecule-1 (NELL-1) has been reported as osteoinductive in multiple in vivo preclinical models. Bone morphogenetic protein (BMP)-2 is used clinically for skeletal repair, but in vivo administration can induce abnormal, adipose-filled, poor-quality bone. We demonstrate that NELL-1 combined with BMP2 significantly optimizes osteogenesis in a rodent femoral segmental defect model by minimizing the formation of BMP2-induced adipose-filled cystlike bone. In vitro studies using the mouse bone marrow stromal cell line M2-10B4 and human primary bone marrow stromal cells have confirmed that NELL-1 enhances BMP2-induced osteogenesis and inhibits BMP2-induced adipogenesis. Importantly, the ability of NELL-1 to direct BMP2-treated cells toward osteogenesis and away from adipogenesis requires intact canonical Wnt signaling. Overall, these studies establish the feasibility of combining NELL-1 with BMP2 to improve clinical bone regeneration and provide mechanistic insight into canonical Wnt pathway activity during NELL-1 and BMP2 osteogenesis. The novel abilities of NELL-1 to stimulate Wnt signaling and to repress adipogenesis may highlight new treatment approaches for bone loss in osteoporosis.


International Orthopaedics | 2014

Roles of bone morphogenetic protein signaling in osteosarcoma

Alan Nguyen; Michelle A. Scott; Sarah M. Dry; Aaron W. James

PurposeSince the original extraction of bone morphogenetic proteins (BMPs) from bovine bone, research interest and clinical use has increased exponentially. With this, a concomitant analysis of BMP expression in bone tumours has been performed. BMP ligands, receptors, and signaling activity have been observed in diverse benign and malignant bone tumours. However, the reported expression, function, and importance of BMPs in bone tumours, and specifically osteosarcomas, have been far from uniform. This review highlights recent advances in understanding the role of BMP signaling in osteosarcoma biology, focusing on the sometimes divergent findings by various researchers and the challenges inherent in the study of osteosarcoma.MethodsWe performed a literature review of all studies examining BMP signaling in osteosarcoma.ResultsOverall, multiple BMP ligands and receptors are expressed in most osteosarcoma cell lines and subtypes, although BMP signaling may be reduced in comparison with benign bone-forming tumours. Studies suggest that osteosarcomas with different lineages of differentiation may have differential expression of BMP ligands. Although significant disagreement in the literature exists, the presence of BMP signaling in osteosarcoma may impart a worse prognosis. On the cellular level, BMP signaling appears to mediate promigratory effects in osteosarcoma and chondrosarcoma cell types, possibly via interaction and activation of Integrin β1.ConclusionsBMP signaling has clear biologic importance in osteosarcoma, although it is not yet fully understood. Future questions for study include assessing the utility of BMP signaling in prognostication of osteosarcoma and the potential modulation of BMP signaling for inhibition of osteosarcomagenesis, growth and invasion.


JCI insight | 2017

NELL-1 induces Sca-1+ mesenchymal progenitor cell expansion in models of bone maintenance and repair

Aaron W. James; Jia Shen; Rebecca Tsuei; Alan Nguyen; Kevork Khadarian; Carolyn A. Meyers; Hsin Chuan Pan; Weiming Li; Jin H. Kwak; Greg Asatrian; Cymbeline T. Culiat; Min Lee; Kang Ting; Xinli Zhang; Chia Soo

NELL-1 is a secreted, osteogenic protein first discovered to control ossification of the cranial skeleton. Recently, NELL-1 has been implicated in bone maintenance. However, the cellular determinants of NELL-1s bone-forming effects are still unknown. Here, recombinant human NELL-1 (rhNELL-1) implantation was examined in a clinically relevant nonhuman primate lumbar spinal fusion model. Prolonged rhNELL-1 protein release was achieved using an apatite-coated β-tricalcium phosphate carrier, resulting in a local influx of stem cell antigen-1-positive (Sca-1+) mesenchymal progenitor cells (MPCs), and complete osseous fusion across all samples (100% spinal fusion rate). Murine studies revealed that Nell-1 haploinsufficiency results in marked reductions in the numbers of Sca-1+CD45-CD31- bone marrow MPCs associated with low bone mass. Conversely, rhNELL-1 systemic administration in mice showed a marked anabolic effect accompanied by increased numbers of Sca-1+CD45-CD31- bone marrow MPCs. Mechanistically, rhNELL-1 induces Sca-1 transcription among MPCs, in a process requiring intact Wnt/β-catenin signaling. In summary, NELL-1 effectively induces bone formation across small and large animal models either via local implantation or intravenous delivery. NELL-1 induces an expansion of a bone marrow subset of MPCs with Sca-1 expression. These findings provide compelling justification for the clinical translation of a NELL-1-based therapy for local or systemic bone formation.


American Journal of Pathology | 2017

The Effects of Systemic Therapy of PEGylated NELL-1 on Fracture Healing in Mice

Justine Tanjaya; Elizabeth L. Lord; Chenchao Wang; Yulong Zhang; Jong Kil Kim; Alan Nguyen; Llyod Baik; Hsin Chuan Pan; Eric Chen; Jin Hee Kwak; Xinli Zhang; Benjamin M. Wu; Chia Soo; Kang Ting

Fractures are common, with an incidence of 13.7 per 1000 adults annually. Systemic agents have been widely used for enhancing bone regeneration; however, the efficacy of these therapeutics for the management and prevention of fracture remains unclear. NEL-like protein 1 (NELL-1) is a potent pro-osteogenic cytokine that has been modified with polyethylene glycol (PEG)ylation [PEGylated NELL-1 (NELL-PEG)] to enhance its pharmacokinetics for systemic therapy. Our aim was to investigate the effects of systemic administration of NELL-PEG on fracture healing in mice and on overall bone properties in uninjured bones. Ten-week-old CD-1 mice were subjected to an open osteotomy of bilateral radii and treated with weekly injections of NELL-PEG or PEG phosphate-buffered saline as control. Systemic injection of NELL-PEG resulted in improved bone mineral density of the fracture site and accelerated callus union. After 4 weeks of treatment, mice treated with NELL-PEG exhibited substantially enhanced callus volume, callus mineralization, and biomechanical properties. NELL-PEG injection significantly augmented bone regeneration, as confirmed by high expression of bone turnover rate, bone formation rate, and mineral apposition rate. Consistently, the immunohistochemistry results also confirmed a high bone remodeling activity in the NELL-PEG-treated group. Our findings suggest that weekly injection of NELL-PEG may have the clinical potential to accelerate fracture union and enhance overall bone properties, which may help prevent subsequent fractures.

Collaboration


Dive into the Alan Nguyen's collaboration.

Top Co-Authors

Avatar

Aaron W. James

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Michelle A. Scott

Nationwide Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Vi Nguyen

University of California

View shared research outputs
Top Co-Authors

Avatar

Chia Soo

University of California

View shared research outputs
Top Co-Authors

Avatar

Jia Shen

University of California

View shared research outputs
Top Co-Authors

Avatar

Kang Ting

University of California

View shared research outputs
Top Co-Authors

Avatar

Dalton Pham

University of California

View shared research outputs
Top Co-Authors

Avatar

Xinli Zhang

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Greg Asatrian

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge