Network


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

Hotspot


Dive into the research topics where Patricia A. Zuk is active.

Publication


Featured researches published by Patricia A. Zuk.


Tissue Engineering | 2001

Multilineage Cells from Human Adipose Tissue: Implications for Cell-Based Therapies

Patricia A. Zuk; Min Zhu; Hiroshi Mizuno; Jerry I. Huang; Futrell Jw; Katz Aj; Prosper Benhaim; Lorenz Hp; Marc H. Hedrick

Future cell-based therapies such as tissue engineering will benefit from a source of autologous pluripotent stem cells. For mesodermal tissue engineering, one such source of cells is the bone marrow stroma. The bone marrow compartment contains several cell populations, including mesenchymal stem cells (MSCs) that are capable of differentiating into adipogenic, osteogenic, chondrogenic, and myogenic cells. However, autologous bone marrow procurement has potential limitations. An alternate source of autologous adult stem cells that is obtainable in large quantities, under local anesthesia, with minimal discomfort would be advantageous. In this study, we determined if a population of stem cells could be isolated from human adipose tissue. Human adipose tissue, obtained by suction-assisted lipectomy (i.e., liposuction), was processed to obtain a fibroblast-like population of cells or a processed lipoaspirate (PLA). These PLA cells can be maintained in vitro for extended periods with stable population doubling and low levels of senescence. Immunofluorescence and flow cytometry show that the majority of PLA cells are of mesodermal or mesenchymal origin with low levels of contaminating pericytes, endothelial cells, and smooth muscle cells. Finally, PLA cells differentiate in vitro into adipogenic, chondrogenic, myogenic, and osteogenic cells in the presence of lineage-specific induction factors. In conclusion, the data support the hypothesis that a human lipoaspirate contains multipotent cells and may represent an alternative stem cell source to bone marrow-derived MSCs.


Cells Tissues Organs | 2003

Comparison of Multi-Lineage Cells from Human Adipose Tissue and Bone Marrow

Daniel A. De Ugarte; Kouki Morizono; Amir Elbarbary; Zeni Alfonso; Patricia A. Zuk; Min Zhu; Jason L. Dragoo; Peter Ashjian; Bert Thomas; Prosper Benhaim; Irvin S. Y. Chen; John K. Fraser; Marc H. Hedrick

Our laboratory has recently characterized a population of cells from adipose tissue, termed processed lipoaspirate (PLA) cells, which have multi-lineage potential similar to bone-marrow-derived mesenchymal stem cells (MSCs). This study is the first comparison of PLA cells and MSCs isolated from the same patient. No significant differences were observed for yield of adherent stromal cells, growth kinetics, cell senescence, multi-lineage differentiation capacity, and gene transduction efficiency. Adipose tissue is an abundant and easily procured source of PLA cells, which have a potential like MSCs for use in tissue-engineering applications and as gene delivery vehicles.


Immunology Letters | 2003

Differential expression of stem cell mobilization-associated molecules on multi-lineage cells from adipose tissue and bone marrow

Daniel A. De Ugarte; Zeni Alfonso; Patricia A. Zuk; Amir Elbarbary; Min Zhu; Peter Ashjian; Prosper Benhaim; Mare H. Hedrick; John K. Fraser

Our laboratory has characterized a population of stromal cells obtained from adipose tissue termed processed lipoaspirate cells (PLAs). PLAs, like bone-marrow derived mesenchymal stem cells (BM-MSCs), have the capacity to differentiate along the adipogenic, osteogenic, chondrogenic, and myogenic lineages, In order to better characterize these two multi-lineage populations, we examined the surface phenotype of both bone marrow and adipose tissue-derived cells from five patients undergoing surgery. PLA and BM-MSC cells were isolated, subcultivated, and evaluated for cell surface marker expression using flow cytometry. PLA and BM-MSC cells both expressed CD13, CD29, CD44, CD90, CD105, SH-3, and STRO-1. Differences in expression were noted for cell adhesion molecules CD49d (Integrin alpha4), CD54 (ICAM-1), CD34, and CD106 (VCAM-1). While markedly similar, the surface phenotypes of PLA and BM-MSC cells are distinct for several cell adhesion molecules implicated in hematopoietic stem cell homing, mobilization, and proliferation.


Plastic and Reconstructive Surgery | 2002

Myogenic differentiation by human processed lipoaspirate cells.

Hiroshi Mizuno; Patricia A. Zuk; Min Zhu; H. Peter Lorenz; Prosper Benhaim; Marc H. Hedrick; James Chang; Anna A. Kuang

&NA; The use of undifferentiated cells for cell‐based tissue engineering and regeneration strategies represents a promising approach for skeletal muscle repair. For such strategies to succeed, a readily available source of myogenic precursor cells must be identified. We have previously shown that cells isolated from raw human lipoaspirates, called processed lipoaspirate cells, display multilineage mesodermal potential in vitro. Because human liposuctioned fat is available in large quantities and can be harvested with low morbidity, it may be an ideal source of stem cells for tissue‐engineering applications. In this study, processed lipoaspirate cells were isolated from raw lipoaspirates harvested from eight patients who underwent cosmetic surgery. Processed lipoaspirate cells were placed in promyogenic conditions for up to 6 weeks, and the expression of the myogenic markers MyoD1 and myosin heavy chain was confirmed by using structure, histology, and reverse transcriptase‐polymerase chain reaction. Histologic results were quantitated as an indicator or myogenic differentiation levels. We found that induced human processed lipoaspirate cells form multinucleated cells after 3 weeks of induction, indicative of the formation of myotubes. In addition, MyoD1 and skeletal muscle myosin heavy chain are expressed at distinct time points during differentiation with MyoD1 expression preceding expression of myosin. Finally, approximately 15 percent of human processed lipoaspirate cells can be induced toward myogenic differentiation 6 weeks after induction. In summary, our findings suggest that human processed lipoaspirate cells differentiate into myogenic cells. Furthermore, these cells may be a useful source for skeletal muscle engineering and repair.


Journal of Orthopaedic Research | 2003

Bone induction by BMP-2 transduced stem cells derived from human fat.

Jason L. Dragoo; Joon Y. Choi; Jay R. Lieberman; Jerry I. Huang; Patricia A. Zuk; Jeffery Zhang; Marc H. Hedrick; Prosper Benhaim

Purpose: We have isolated pluripotent mesenchymal progenitor cells in large numbers from liposuction aspirates (processed lipoaspirate cells or PLAs). This study examines the osteogenic potential of PLAs and bone marrow aspirate cells (BMAs), when exposed to either recombinant human bone morphogenetic protein (BMP)‐2 (rh‐BMP‐2) or adenovirus containing BMP‐2 cDNA (Ad‐BMP‐2).


Plastic and Reconstructive Surgery | 2003

IN VITRO DIFFERENTIATION OF HUMAN PROCESSED LIPOASPIRATE CELLS INTO EARLY NEURAL PROGENITORS

Peter Ashjian; Amir Elbarbary; Brian Edmonds; Daniel A. DeUgarte; Min Zhu; Patricia A. Zuk; H. Peter Lorenz; Prosper Benhaim; Marc H. Hedrick

Human processed lipoaspirate (PLA) cells are multipotent stem cells, capable of differentiating into multiple mesenchymal lineages (bone, cartilage, fat, and muscle). To date, differentiation to nonmesodermal fates has not been reported. This study demonstrates that PLA cells can be induced to differentiate into early neural progenitors, which are of an ectodermal origin. Undifferentiated cultures of human PLA cells expressed markers characteristic of neural cells such as neuron-specific enolase (NSE), vimentin, and neuron-specific nuclear protein (NeuN). After 2 weeks of treatment of PLA cells with isobutylmethylxanthine, indomethacin, and insulin, about 20 to 25 percent of the cells differentiated into cells with typical neural morphologic characteristics, accompanied by increased expression of NSE, vimentin, and the nerve-growth factor receptor trk-A. However, induced PLA cells did not express the mature neuronal marker, MAP, or the mature astrocyte marker, GFAP. It was also found that neurally induced PLA cells displayed a delayed-rectifier type K+ current (an early developmental ion channel) concomitantly with morphologic changes and increased expression of neural-specific markers. The authors concluded that human PLA cells might have the potential to differentiate in vitro into cells that represent early progenitors of neurons and/or glia.


Molecular Biology of the Cell | 2010

The Adipose-derived Stem Cell: Looking Back and Looking Ahead

Patricia A. Zuk

In 2002, researchers at UCLA published a manuscript in Molecular Biology of the Cell describing a novel adult stem cell population isolated from adipose tissue—the adipose-derived stem cell (ASC). Since that time, the ASC has gone on to be one of the most popular adult stem cell populations currently being used in the stem cell field. With multilineage mesodermal potential and possible ectodermal and endodermal potentials also, the ASC could conceivably be an alternate to pluripotent ES cells in both the lab and in the clinic. In this retrospective article, a historical perspective on the ASC is given together with exciting new applications for the stem cell being considered today.


Plastic and Reconstructive Surgery | 2004

Chondrogenic potential of multipotential cells from human adipose tissue

Jerry I. Huang; Patricia A. Zuk; Neil F. Jones; Min Zhu; H. Peter Lorenz; Marc H. Hedrick; Prosper Benhaim

The use of stem cells for cell-based tissue-engineering strategies represents a promising alternative for the repair of cartilaginous defects. The multilineage potential of a population of putative mesodermal stem cells obtained from human lipoaspirates, termed processed lipoaspirate cells, was previously characterized. The chondrogenic potential of those cells was confirmed with a combination of histological and molecular approaches. Processed lipoaspirate cells under high-density micromass culture conditions, supplemented with transforming growth factor-&bgr;1, insulin, transferrin, and ascorbic acid, formed well-defined nodules within 48 hours of induction and expressed the cartilaginous markers collagen type II, chondroitin-4-sulfate, and keratan sulfate. Reverse transcription polymerase chain reaction analysis confirmed the expression of collagen type II and the cartilage-specific proteoglycan aggrecan. In summary, human adipose tissue may represent a novel plentiful source of multipotential stem cells capable of undergoing chondrogenesis in vitro.


Journal of Tissue Engineering and Regenerative Medicine | 2009

The effect of age on osteogenic, adipogenic and proliferative potential of female adipose-derived stem cells.

Min Zhu; Emil Kohan; James P. Bradley; Marc H. Hedrick; Prosper Benhaim; Patricia A. Zuk

Human adipose tissue is an ideal source of autologous cells that is both plentiful and easily obtainable in large quantities through the simple surgical procedure of liposuction. The stromal vascular fraction of adipose tissue contains a stem cell population, adipose‐derived stem cells (ASCs), capable of adipogenic, osteogenic, myogenic and chondrogenic differentiation. These cells have already been recognized to possess great therapeutic potential in tissue engineering and regeneration. In this study, we sought to determine the effect of donor age on the growth kinetics and differentiation potential of ASCs. For this, ASCs were isolated from liposuctioned adipose tissue obtained from female patients in the age range 20–58 years. Population doubling time was calculated over 2 weeks and differentiation potential was determined by assaying for adipogenesis and osteogenesis. ASCs obtained from older donors appeared to have a slower rate of proliferation, but this relationship was not significant. While adipogenic potential was unrelated to donor age, a distinct relationship between donor age and osteogenic potential was observed. The aetiology of this age‐dependent change in osteogenic potential was not due to any changes in the number of precursors with osteogenic capacity in the adipose sample. These findings have important implications for emerging cell‐based therapeutic strategies, such as tissue engineering, in addition to treatment of various metabolic bone disorders including osteoporosis. Copyright


Cells Tissues Organs | 2008

Human Adipose Stem Cells: A Potential Cell Source for Cardiovascular Tissue Engineering

Sepideh Heydarkhan-Hagvall; Katja Schenke-Layland; Jin Q. Yang; Sanaz Heydarkhan; Yuhuan Xu; Patricia A. Zuk; W. Robb MacLellan; Ramin E. Beygui

Background/Aims: A crucial step in providing clinically relevant applications of cardiovascular tissue engineering involves the identification of a suitable cell source. The objective of this study was to identify the exogenous and endogenous parameters that are critical for the differentiation of human adipose stem cells (hASCs) into cardiovascular cells. Methods: hASCs were isolated from human lipoaspirate samples, analyzed, and subjected to two differentiation protocols. Results: As shown by fluorescence-activated cell sorter (FACS) analysis, a population of hASCs expressed stem cell markers including CXCR4, CD34, c-kit, and ABCG2. Further, FACS and immunofluorescence analysis of hASCs, cultured for 2 weeks in DMEM-20%-FBS, showed the expression of smooth muscle cell (SMC)-specific markers including SM α-actin, basic calponin, h-caldesmon and SM myosin. hASCs, cultured for 2 weeks in endothelial cell growth medium-2 (EGM-2), formed a network of branched tube-like structures positive for CD31, CD144, and von Willebrand factor. The frequency of endothelial cell (EC) marker-expressing cells was passage number-dependent. Moreover, hASCs attached and formed a confluent layer on top of electrospun collagen-elastin scaffolds. Scanning electron microscopy and DAPI staining confirmed the integration of hASCs with the fibers and formation of a cell-matrix network. Conclusion: Our results indicate that hASCs are a potential cell source for cardiovascular tissue engineering; however, the differentiation capacity of hASCs into SMCs and ECs is passage number- and culture condition-dependent.

Collaboration


Dive into the Patricia A. Zuk's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Reza Jarrahy

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Min Zhu

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Benjamin M. Wu

University of California

View shared research outputs
Top Co-Authors

Avatar

Andrew J. Li

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Peter Ashjian

University of California

View shared research outputs
Top Co-Authors

Avatar

Sarah Sorice

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge