scispace - formally typeset
Search or ask a question
Author

Peng Huang

Bio: Peng Huang is an academic researcher from Mayo Clinic. The author has contributed to research in topics: Mesenchymal stem cell & Stromal cell. The author has an hindex of 5, co-authored 7 publications receiving 140 citations.

Papers
More filters
Journal ArticleDOI
05 Aug 2013-PLOS ONE
TL;DR: POM is a promising therapeutic agent for CNS lymphoma with excellent CNS penetration, significant preclinical therapeutic activity, and a major impact on the tumor microenvironment.
Abstract: Primary CNS lymphoma carries a poor prognosis. Novel therapeutic agents are urgently needed. Pomalidomide (POM) is a novel immunomodulatory drug with anti-lymphoma activity. CNS pharmacokinetic analysis was performed in rats to assess the CNS penetration of POM. Preclinical evaluation of POM was performed in two murine models to assess its therapeutic activity against CNS lymphoma. The impact of POM on the CNS lymphoma immune microenvironment was evaluated by immunohistochemistry and immunofluorescence. In vitro cell culture experiments were carried out to further investigate the impact of POM on the biology of macrophages. POM crosses the blood brain barrier with CNS penetration of ~ 39%. Preclinical evaluations showed that it had significant therapeutic activity against CNS lymphoma with significant reduction in tumor growth rate and prolongation of survival, that it had a major impact on the tumor microenvironment with an increase in macrophages and natural killer cells, and that it decreased M2-polarized tumor-associated macrophages and increased M1-polarized macrophages when macrophages were evaluated based on polarization status. In vitro studies using various macrophage models showed that POM converted the polarization status of IL4-stimulated macrophages from M2 to M1, that M2 to M1 conversion by POM in the polarization status of lymphoma-associated macrophages is dependent on the presence of NK cells, that POM induced M2 to M1 conversion in the polarization of macrophages by inactivating STAT6 signaling and activating STAT1 signaling, and that POM functionally increased the phagocytic activity of macrophages. Based on our findings, POM is a promising therapeutic agent for CNS lymphoma with excellent CNS penetration, significant preclinical therapeutic activity, and a major impact on the tumor microenvironment. It can induce significant biological changes in tumor-associated macrophages, which likely play a major role in its therapeutic activity against CNS lymphoma. POM should be further evaluated in clinical trials.

74 citations

Journal ArticleDOI
TL;DR: It was shown that M17 proliferation was significantly decreased and the rate of apoptosis increased after exposure to OGD, and these effects could be alleviated via co-culture with MSCs.

56 citations

Journal ArticleDOI
TL;DR: Intraventricular administration of MSCs in ICH rat model showed improved behavior and alleviated brain damage, and treated ICH rats showed significantly reduced expression of IL-1α, IL-6, and IFN-γ.
Abstract: External ventricular drain (EVD) is used clinically to relieve intracranial pressure and occasionally to deliver medications following intracerebral hemorrhage (ICH) Mesenchymal stem cell (MSC) therapy has been shown to be neuroprotective and can induce neuroregeneration in stroke models We evaluated the safety and efficacy of delivering MSCs intraventricularly in a rat hemorrhagic stroke model Using autologous blood, hemorrhagic stroke was induced at specific coordinates in the right basal ganglia After 30 minutes, rats were treated with either bone marrow-derived MSCs or a phosphate-buffered saline placebo via direct intraventricular infusion Three dosages (2 × 105/kg, 5 × 105/kg, and 1 × 106/kg) of MSCs were administered Forelimb use asymmetry test was employed to evaluate functional improvement after cell therapy At the end of the experiment, peripheral blood samples and organs were harvested; biochemistry, cytokine, and growth factor analysis and histology evaluations were performed to explore cell toxicity and cell fate, and the effects of MSC therapy on injury volume, anti-inflammation, and neurogenesis Intraventricular administration of MSCs in ICH rat model showed improved behavior and alleviated brain damage Additionally, treated ICH rats showed significantly reduced expression of IL-1α, IL-6, and IFN-γ No obvious cell toxicity was noticed through blood chemistry and histology evaluation None of the infused MSCs were detected at the end of the experiment EVD is safe and effective to use as a method of delivering MSCs to treat ICH Intraventricularly delivered MSCs have anti-inflammatory properties and a capacity to induce neurogenesis and improve function following ICH injury

38 citations

Journal ArticleDOI
01 Jun 2020
TL;DR: It was found that microgravity had significant impact on the MSC capacity to secrete cytokines and growth factors, and appeared to be more potent in terms of immunosuppressive capacity compared to their identical ground control.
Abstract: Growing stem cells on Earth is very challenging and limited to a few population doublings. The standard two-dimensional (2D) culture environment is an unnatural condition for cell growth. Therefore, culturing stem cells aboard the International Space Station (ISS) under a microgravity environment may provide a more natural three-dimensional environment for stem cell expansion and organ development. In this study, human-derived mesenchymal stem cells (MSCs) grown in space were evaluated to determine their potential use for future clinical applications on Earth and during long-term spaceflight. MSCs were flown in Plate Habitats for transportation to the ISS. The MSCs were imaged every 24–48 h and harvested at 7 and 14 days. Conditioned media samples were frozen at −80 °C and cells were either cryopreserved in 5% dimethyl sulfoxide, RNAprotect, or paraformaldehyde. After return to Earth, MSCs were characterized to establish their identity and cell cycle status. In addition, cell proliferation, differentiation, cytokines, and growth factors’ secretion were assessed. To evaluate the risk of malignant transformation, the space-grown MSCs were subjected to chromosomal, DNA damage, and tumorigenicity assays. We found that microgravity had significant impact on the MSC capacity to secrete cytokines and growth factors. They appeared to be more potent in terms of immunosuppressive capacity compared to their identical ground control. Chromosomal, DNA damage, and tumorigenicity assays showed no evidence of malignant transformation. Therefore, it is feasible and potentially safe to grow MSCs aboard the ISS for potential future clinical applications.

22 citations

Journal ArticleDOI
10 Nov 2021-Cells
TL;DR: In this paper, the authors evaluated the feasibility of transfecting IL-10 mRNA to enhance MSC therapeutic potential, and showed that IL10 mRNA transfection did not appear to alter MSC intrinsic characteristics.
Abstract: Mesenchymal stem cells (MSCs) are used in various studies to induce immunomodulatory effects in clinical conditions associated with immune dysregulation such as graft versus host disease (GvHD). However, most of these clinical trials failed to go beyond early phase 2 studies because of limited efficacy. Various methods have been assessed to increase the potency of MSCs. IL-10 is an anti-inflammatory cytokine that is known to modulate immune responses in GvHD. In this study, we evaluated the feasibility of transfecting IL-10 mRNA to enhance MSC therapeutic potential. IL-10 mRNA engineered MSCs (eMSCs-IL10) maintained high levels of IL-10 expression even after freezing and thawing. IL-10 mRNA transfection did not appear to alter MSC intrinsic characteristics. eMSCs-IL10 significantly suppressed T cell proliferation relative to naive MSCs in vitro. In a mouse model for GvHD, eMSCs-IL10 induced a decrease in plasma level of potent pro-inflammatory cytokines and inhibited CD4+ and CD8+ T cell proliferation in the spleen. In summary, our studies demonstrate the feasibility of potentiating MSCs to enhance their immunomodulatory effects by IL-10 mRNA transfection. The use of non-viral transfection may generate a safe and potent MSC product for treatment of clinical conditions associated with immune dysregulation such as GvHD.

9 citations


Cited by
More filters
01 Jan 1980

1,523 citations

Journal ArticleDOI
TL;DR: Experimental studies of ischemic stroke showed that EVs have immunomodulatory and neuroprotective properties; therefore, they can stimulate neurogenesis and angiogenesis and have the capacity to cross the blood-brain barrier.
Abstract: Ischemic stroke is the third cause of death in the developed countries and the main reason of severe disability. Brain ischemia leads to the production of damage-associated molecular patterns (DAMPs) by neurons and glial cells which results in astrocyte and microglia activation, pro-inflammatory cytokines and chemokines production, blood-brain barrier (BBB) disruption, infiltration of leukocytes from the peripheral blood into the infarcted area, and further exacerbation of tissue damage. However, some immune cells such as microglia or monocytes are capable to change their phenotype to anti-inflammatory, produce anti-inflammatory cytokines, and protect injured nervous tissue. In this situation, therapies, which will modulate the immune response after brain ischemia, such as transplantation of mesenchymal stem cells (MSCs) are catching interest. Many experimental studies of ischemic stroke revealed that MSCs are able to modulate immune response and act neuroprotective, through stimulation of neurogenesis, oligodendrogenesis, astrogenesis, and angiogenesis. MSCs may also have an ability to replace injured cells, but the release of paracrine factors directly into the environment or via extracellular vesicles (EVs) seems to play the most pronounced role. EVs are membrane structures containing proteins, lipids, and nucleic acids, and they express similar properties as the cells from which they are derived. However, EVs have lower immunogenicity, do not express the risk of vessel blockage, and have the capacity to cross the blood-brain barrier. Experimental studies of ischemic stroke showed that EVs have immunomodulatory and neuroprotective properties; therefore, they can stimulate neurogenesis and angiogenesis. Up to now, 20 clinical trials with MSC transplantation into patients after stroke were performed, from which two concerned on only hemorrhagic stroke and 13 studied only on ischemic stroke. There is no clinical trial with EV injection into patients after brain ischemia so far, but the case with miR-124-enriched EVs administration is planned and probably there will be more clinical studies with EV transplantation in the near future.

171 citations

Journal ArticleDOI
23 Jan 2014-Blood
TL;DR: The incidence, clinical presentation, and prognostic factors of PTL are discussed and a summary of the recent advances in the authors' understanding of its pathophysiology are presented, which may account for the characteristic clinical features.

160 citations

Journal ArticleDOI
25 Feb 2016-Blood
TL;DR: The present review uses a case-and-comment format to describe the approach to the management of EMM, which shows that EMM clinical situations are extraordinarily heterogeneous, and their management is particularly challenging.

122 citations

Journal ArticleDOI
22 Nov 2018-Blood
TL;DR: POM/DEX treatment is feasible with significant therapeutic activity against relapsed/refractory PCNSL and PVRL and overall response rate (ORR), progression-free survival (PFS), and safety profile as secondary objectives.

82 citations