Network


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

Hotspot


Dive into the research topics where Ching-Shwun Lin is active.

Publication


Featured researches published by Ching-Shwun Lin.


Stem Cells and Development | 2008

Defining stem and progenitor cells within adipose tissue.

Guiting Lin; Maurice Garcia; Hongxiu Ning; Lia Banie; Yinglu Guo; Tom F. Lue; Ching-Shwun Lin

Adipose tissue-derived stem cells (ADSC) are routinely isolated from the stromal vascular fraction (SVF) of homogenized adipose tissue. Freshly isolated ADSC display surface markers that differ from those of cultured ADSC, but both cell preparations are capable of multipotential differentiation. Recent studies have inferred that these progenitors may reside in a perivascular location where they appeared to coexpress CD34 and smooth muscle actin (alpha-SMA) but not CD31. However, these studies provided only limited histological evidence to support such assertions. In the present study, we employed immunohistochemistry and immunofluorescence to define more precisely the location of ADSC within human adipose tissue. Our results show that alpha-SMA and CD31 localized within smooth muscle and endothelial cells, respectively, in all blood vessels examined. CD34 localized to both the intima (endothelium) and adventitia neither of which expressed alpha-SMA. The niche marker Wnt5a was confined exclusively to the vascular wall within mural smooth muscle cells. Surprisingly, the widely accepted mesenchymal stem cell marker STRO-1 was expressed exclusively in the endothelium of capillaries and arterioles but not in the endothelium of arteries. The embryonic stem cell marker SSEA1 localized to a pericytic location in capillaries and in certain smooth muscle cells of arterioles. Cells expressing the embryonic stem cell markers telomerase and OCT4 were rare and observed only in capillaries. Based on these findings and evidence gathered from the existing literature, we propose that ADSC are vascular precursor (stem) cells at various stages of differentiation. In their native tissue, ADSC at early stages of differentiation can differentiate into tissue-specific cells such as adipocytes. Isolated, ADSC can be induced to differentiate into additional cell types such as osteoblasts and chondrocytes.


Histology and Histopathology | 2010

Defining adipose tissue-derived stem cells in tissue and in culture.

Ching-Shwun Lin; Zhongcheng Xin; Chunhua Deng; Hongxiu Ning; Guitin Lin; Tom F. Lue

Adipose tissue-derived stem cells (ADSC) are routinely isolated from the stromal vascular fraction (SVF) of homogenized adipose tissue. Similar to other types of mesenchymal stem cells (MSC), ADSC remain difficult to define due to the lack of definitive cellular markers. Still, many types of MSC, including ADSC, have been shown to reside in a perivascular location, and increasing evidence shows that both MSC and ADSC may in fact be vascular stem cells (VSC). Locally, these cells differentiate into smooth muscle and endothelial cells that are assembled into newly formed blood vessels during angiogenesis and neovasculogenesis. Additionally, MSC or ADSC can also differentiate into tissue cells such as adipocytes in the adipose tissue. Systematically, MSC or ADSC are recruited to injury sites where they participate in the repair/regeneration of the injured tissue. Due to the vasculatures dynamic capacity for growth and multipotential nature for diversification, VSC in tissue are individually at various stages and on different paths of differentiation. Therefore, when isolated and put in culture, these cells are expected to be heterogeneous in marker expression, renewal capacity, and differentiation potential. Although this heterogeneity of VSC does impose difficulties and cause confusions in basic science studies, its impact on the development of VSC as a therapeutic cell source has not been as apparent, as many preclinical and clinical trials have reported favorable outcomes. With this understanding, ADSC are generally defined as CD34+CD31- although loss of CD34 expression in culture is well documented. In adipose tissue, CD34 is localized to the intima and adventitia of blood vessels but not the media where cells expressing alpha-smooth muscle actin (SMA) exist. By excluding the intima, which contains the CD34+CD31+ endothelial cells, and the media, which contains the CD34-CD31- smooth muscle cells, it leaves the adventitia as the only possible location for the CD34+ ADSC. In the capillary, CD34 and CD140b (a pericyte marker) are mutually exclusively expressed, thus suggesting that pericytes are not the CD34+ ADSC. Many other cellular markers for vascular cells, stem cells, and stem cell niche have also been investigated as possible ADSC markers. Particularly the best-known MSC marker STRO-1 has been found either expressed or not expressed in cultured ADSC. In the adipose tissue, STRO-1 appears to be expressed exclusively in the endothelium of certain but not all blood vessels, and thus not associated with the CD34+ ADSC. In conclusion, we believe that ADSC exist as CD34+CD31-CD104b-SMA- cells in the capillary and in the adventitia of larger vessels. In the capillary these cells coexist with pericytes and endothelial cells, both of which are possibly progenies of ADSC (or more precisely VSC). In the larger vessels, these ADSC or VSC exist as specialized fibroblasts (having stem cell properties) in the adventitia.


The Journal of Sexual Medicine | 2010

Injections of Adipose Tissue-Derived Stem Cells and Stem Cell Lysate Improve Recovery of Erectile Function in a Rat Model of Cavernous Nerve Injury

Maarten Albersen; Thomas M. Fandel; Guiting Lin; Guifang Wang; Lia Banie; Ching-Shwun Lin; Tom F. Lue

INTRODUCTION Erectile dysfunction (ED) remains a major complication after radical prostatectomy. The use of adipose tissue-derived stem cells (ADSCs) has shown promising results for the treatment of ED. However, the mechanisms of action for stem cell therapy remain controversial, with increasing evidence pointing to paracrine pathways. AIM To determine the effects and to identify the mechanism of action of ADSC and ADSC-derived lysate in a rat model of cavernous nerve (CN) crush injury. METHODS Thirty-two male Sprague-Dawley rats were randomly divided into four equal groups: one group underwent sham operation, while three groups underwent bilateral CN crush. Crush-injury groups were treated at the time of injury with intracavernous injection of ADSC, lysate, or vehicle only (injured controls). Erectile function was assessed by CN electrostimulation at 4 weeks. Penile tissue was collected for histology. MAIN OUTCOME MEASURES   Intracavernous pressure increase upon CN stimulation; neuronal nitric oxide synthase (nNOS) content in the dorsal penile nerve; smooth muscle content, collagen content, and number of apoptotic cells in the corpus cavernosum. RESULTS Both ADSC and lysate treatments resulted in significant recovery of erectile function, as compared with vehicle treatment. nNOS content was preserved in both the ADSC and lysate group, with significantly higher expression compared with vehicle-treated animals. There was significantly less fibrosis and a significant preservation of smooth muscle content in the ADSC and lysate groups compared with injured controls. The observed functional improvement after lysate injection supports the hypothesis that ADSCs act through release of intracellular preformed substances or by active secretion of certain biomolecules. The underlying mechanism of recovery appears to involve neuron preservation and cytoprotection by inhibition of apoptosis. CONCLUSIONS Penile injection of both ADSC and ADSC-derived lysate can improve recovery of erectile function in a rat model of neurogenic ED.


International Journal of Impotence Research | 2003

Intracavernosal vascular endothelial growth factor (VEGF) injection and adeno-associated virus-mediated VEGF gene therapy prevent and reverse venogenic erectile dysfunction in rats

Rogers Rs; Graziottin Tm; Ching-Shwun Lin; Kan Yw; Tom F. Lue

Penile veno-occlusive dysfunction (venogenic erectile dysfunction) is a common cause of erectile dysfunction (ED). We investigated whether vascular endothelial growth factor (VEGF) can be used to prevent and reverse venogenic ED in a rat model. Pharmacological cavernosometry was developed and validated using adult male rats with either arteriogenic or venogenic ED. Castrated animals were treated with intracavernous VEGF as either a recombinant protein (C+VEGF) or adeno-associated virus (AAV)-mediated VEGF gene therapy (C+VEGF gene) in an attempt to prevent the development of venogenic ED. Other animal groups received testosterone replacement (C+testosterone) or intracavernous AAV-LacZ gene (C+LacZ). Animals with documented venogenic ED were treated with intracavernous VEGF in an attempt to reverse their ED. Functional analysis (pharmacological infusion cavernosometry) was performed following treatment. Penile specimens were harvested for immunohistochemistry and electron microscopic evaluation. Castrated rats showed a decrease in papaverine-induced intracavernous pressure and an increase in maintenance and drop rates during pharmacological cavernosometry. These changes were prevented by systemic testosterone and intracavernous VEGF or AAV-VEGF therapy. Moreover, intracavernous VEGF was able to reverse the venogenic ED produced by castration. The quantity of penile smooth muscle detected by alpha actin staining decreased after castration but not in the C+T, C+VEGF, or C+VEGF gene groups. Transmission electron microscopy revealed atrophy of penile smooth muscle cells and nerves in the castrated rats. In VEGF-treated rats, regeneration of smooth muscle and nerves as well as endothelial cell hypertrophy and hyperplasia were the prominent features. In our animal model, systemic testosterone replacement or intracavernous VEGF (protein and VEGF gene) prevented the veno-occlusive dysfunction in castrated animals. In rats with established venous leakage, VEGF treatment reversed the cavernosometric findings of leakage. Intracavernous injection of either VEGF protein or VEGF gene may be a preferred therapy to preserve erectile function in patients in whom testosterone therapy is contraindicated.


Cytotherapy | 2010

Treatment of stress urinary incontinence with adipose tissue-derived stem cells.

Guiting Lin; Guifang Wang; Lia Banie; Hongxiu Ning; Alan W. Shindel; Thomas M. Fandel; Tom F. Lue; Ching-Shwun Lin

BACKGROUND AIMS Effective treatment for stress urinary incontinence (SUI) is lacking. This study investigated whether transplantation of adipose tissue-derived stem cells (ADSC) can treat SUI in a rat model. METHODS Rats were induced to develop SUI by postpartum vaginal balloon dilation and bilateral ovariectomy. ADSC were isolated from the peri-ovary fat, examined for stem cell properties, and labeled with thymidine analog BrdU or EdU. Ten rats received urethral injection of saline as a control. Twelve rats received urethral injection of EdU-labeled ADSC and six rats received intravenous injection of BrdU-labeled ADSC through the tail vein. Four weeks later, urinary voiding function was assessed by conscious cystometry. The rats were then killed and their urethras harvested for tracking of ADSC and quantification of elastin, collagen and smooth muscle contents. RESULTS Cystometric analysis showed that eight out 10 rats in the control group had abnormal voiding, whereas four of 12 (33.3%) and two of six (33.3%) rats in the urethra-ADSC and tail vein-ADSC groups, respectively, had abnormal voiding. Histologic analysis showed that the ADSC-treated groups had significantly higher elastin content than the control group and, within the ADSC-treated groups, rats with normal voiding pattern also had significantly higher elastin content than rats with voiding dysfunction. ADSC-treated normal-voiding rats had significantly higher smooth muscle content than control or ADSC-treated rats with voiding dysfunction. CONCLUSIONS Transplantation of ADSC via urethral or intravenous injection is effective in the treatment and/or prevention of SUI in a pre-clinical setting.


BJUI | 2004

The effect of neural embryonic stem cell therapy in a rat model of cavernosal nerve injury

Derek Bochinski; G. Lin; Lora Nunes; Rafael Carrion; Nadeem U. Rahman; Ching-Shwun Lin; Tom F. Lue

To isolate embryonic stem cells that have differentiated along the neuronal cell line, and to assess whether injecting these neural stem cells into the corpus cavernosum influences cavernosal nerve regeneration and functional status.


The Journal of Urology | 2001

The effect of adeno-associated virus mediated brain derived neurotrophic factor in an animal model of neurogenic impotence

Mustafa Emre Bakircioglu; Ching-Shwun Lin; Peidong Fan; Karl-Dietrich Sievert; Yeut W. Kan; Tom F. Lue

PURPOSE We tested the hypothesis that transfecting penile tissue with brain derived neurotrophic factor may facilitate neural recovery and erectile capability after cavernous nerve injury. MATERIALS AND METHODS Of the 34 Sprague-Dawley rats used 10 underwent sham operation and 24 underwent bilateral cavernous nerve freezing and intracavernous injection of adeno-associated virus-LacZ (12) or adeno-associated virus-brain derived neurotrophic factor (12). Erectile function was assessed by cavernous nerve electrostimulation at 4 and 8 weeks, and samples of penile tissue and the major pelvic ganglia were evaluated histologically. RESULTS In the brain derived neurotrophic factor group mean maximal intracavernous pressure plus or minus standard deviation was significantly higher than in the LacZ group at 4 and 8 weeks (58.5 +/- 11.7 cm. water versus 28.4 +/- 5.5 and 61.3 +/- 12.5 versus 37.7 +/- 7.9, respectively). In addition, in the brain derived neurotrophic factor group reduced nicotinamide adenine dinucleotide phosphate diaphorase staining and neuronal nitric oxide synthase immunostaining revealed significantly more positive nerve fibers in the dorsal nerves and cavernous tissue than in the LacZ group at each time point and the percent of neuronal nitric oxide synthase positive neurons in the major pelvic ganglia was also significantly greater. Moreover, in the LacZ group most neurons showed a light staining pattern with irregular contours and numerous vacuoles in the cytoplasm. CONCLUSIONS Intracavernous injection of adeno-associated virus-brain derived neurotrophic factor may prevent the degeneration of neuronal nitric oxide synthase containing neurons in the major pelvic ganglia and facilitate the regeneration of neuronal nitric oxide synthase containing nerve fibers in penile tissue, thus, enhancing the recovery of erectile function after bilateral cavernous nerve injury.


The Journal of Sexual Medicine | 2009

Fibroblast Growth Factor 2 Promotes Endothelial Differentiation of Adipose Tissue-Derived Stem Cells

Hongxiu Ning; Gang Liu; Guiting Lin; Rong Yang; Tom F. Lue; Ching-Shwun Lin

INTRODUCTION Adipose tissue-derived stem cells (ADSC) could potentially restore endothelial function in vasculogenic erectile dysfunction (ED). The mechanism for ADSC endothelial differentiation remained unidentified. AIM To test whether ADSC could differentiate into endothelial cells in the penis and to identify the underlying mechanism of ADSC endothelial differentiation. METHODS For in vivo endothelial differentiation, ADSC were labeled with bromodeoxyuridine (BrdU), injected into rat corpora cavernosa, and localized by immunofluorescence and phase-contrast microscopy. For in vitro endothelial differentiation, ADSC were grown in endothelial growth medium 2 (EGM2), stained for endothelial markers CD31, von Willebrand Factor (vWF), and endothelial nitric oxide synthase (eNOS), and assessed for the ability to form tube-like structures in Matrigel and to endocytose acetylated low-density lipoprotein (Ac-LDL). To identify factors that promote ADSC endothelial differentiation, ADSC were grown in various media, each of which contained a specific combination of supplemental factors and assessed for LDL-uptake. PD173074, a selective inhibitor of fibroblast growth factor 2 (FGF2) receptor, was used to confirm the importance of FGF2 signaling for ADSC endothelial differentiation. MAIN OUTCOME MEASURES In vivo endothelial differentiation was assessed by immunofluorescence microscopy. In vitro endothelial differentiation was assessed by immunofluorescence, Matrigel tube formation, and Ac-LDL uptake. RESULTS Injected ADSC were localized to the sinusoid endothelium, some of which stained positive for both BrdU and endothelial antigen rat endothelial cell antigen. ADSC proliferated at a faster rate in EGM2 than in standard DMEM, expressed endothelial markers CD31, vWF, and eNOS, formed tube-like structures in Matrigel, and endocytosed Ac-LDL. These properties were greatly diminished when ADSC were grown in the absence of FGF2 but were unaffected when grown in the absence of vascular endothelial growth factor, insulin-like growth factor, or epidermal growth factor. Furthermore, ADSC displayed similar endothelial properties when grown in FGF2-supplemented basic medium as in EGM2. Finally, blockade of FGF2 signaling with PD173074 abrogated ADSC endothelial differentiation. CONCLUSIONS ADSC could differentiate into endothelial cells in the penis. FGF2 signaling mediates ADSC endothelial differentiation.


Cytotherapy | 2012

Is CD34 truly a negative marker for mesenchymal stromal cells

Ching-Shwun Lin; Hongxiu Ning; Guiting Lin; Tom F. Lue

The prevailing school of thought is that mesenchymal stromal cells (MSC) do not express CD34, and this sets MSC apart from hematopoietic stem cells (HSC), which do express CD34. However, the evidence for MSC being CD34(-) is largely based on cultured MSC, not tissue-resident MSC, and the existence of CD34(-) HSC is in fact well documented. Furthermore, the Stro-1 antibody, which has been used extensively for the identification/isolation of MSC, was generated by using CD34(+) bone marrow cells as immunogen. Thus, neither MSC being CD34(-) nor HSC being CD34(+) is entirely correct. In particular, two studies that analyzed CD34 expression in uncultured human bone marrow nucleated cells found that MSC (BMSC) existed in the CD34(+) fraction. Several studies have also found that freshly isolated adipose-derived MSC (ADSC) express CD34. In addition, all of these ADSC studies and several other MSC studies have observed a disappearance of CD34 expression when the cells are propagated in culture. Thus the available evidence points to CD34 being expressed in tissue-resident MSC, and its negative finding being a consequence of cell culturing.


Current Pharmaceutical Design | 2006

Expression, Distribution and Regulation of Phosphodiesterase 5

Ching-Shwun Lin; Guiting Lin; Zhongcheng Xin; Tom F. Lue

Phosphodiesterase 5 (PDE5) is one of eleven members of the mammalian phosphodiesterase family that hydrolyzes cyclic guanosine monophosphate (cGMP) and cyclic adenosine monophosphate (cAMP). Best known as the target of the impotence drug sildenafil, PDE5 degrades cGMP in smooth muscle cells so as to maintain the contracted state of contractile organs such as the penis, blood vessels, uterus, and intestines. In addition, it regulates numerous other physiological processes such as neurogenesis and apoptosis. Like all other PDEs, PDE5 is dimeric; each subunit is approximately 100 kd in size and has two allosteric cGMP-binding sites and a catalytic domain. Protein kinase G (PKG)-mediated phosphorylation and allosteric cGMP binding upregulate PDE5 activity, while PP1 phosphatase-mediated dephosphorylation downregulates. Sildenafil and other selective inhibitors inhibit PDE5 by binding to the catalytic site. From two promoters a single PDE5A gene at human chromosome 4q26 encodes three alternatively spliced isoforms (PDE5A1-3) that differ in the N-terminus. The PDE5A promoter is located upstream of the three isoform-specific first exons (in the order of A1-A3-A2) and consists of a 139-bp core, a 308-bp upstream enhancer, and a 156-bp downstream enhancer. The weaker 182-bp PDE5A2 promoter is located between the A3- and A2-specific exons and contains an indispensable Sp1-binding sequence. Both promoters are responsive to cGMP or cAMP stimulation, and several studies have demonstrated regulation of PDE5 expression possibly through these promoters. Virtually all tissues and cell types express PDE5, with heart and cardiomyocytes being contentious. PDE5A1 and PDE5A2 are ubiquitous, but PDE5A3 is specific to smooth muscle.

Collaboration


Dive into the Ching-Shwun Lin's collaboration.

Top Co-Authors

Avatar

Tom F. Lue

University of California

View shared research outputs
Top Co-Authors

Avatar

Guiting Lin

University of California

View shared research outputs
Top Co-Authors

Avatar

Hongxiu Ning

University of California

View shared research outputs
Top Co-Authors

Avatar

Lia Banie

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Guifang Wang

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Maarten Albersen

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Xuefeng Qiu

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge