Network


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

Hotspot


Dive into the research topics where Hao Wang is active.

Publication


Featured researches published by Hao Wang.


Journal of Translational Medicine | 2007

Endometrial regenerative cells: A novel stem cell population

Xiaolong Meng; Thomas E. Ichim; Jie Zhong; Andrea Rogers; Zhenglian Yin; James A. Jackson; Hao Wang; Wei Ge; Vladimir Bogin; Kyle Chan; Bernard Thébaud; Neil H. Riordan

Angiogenesis is a critical component of the proliferative endometrial phase of the menstrual cycle. Thus, we hypothesized that a stem cell-like population exist and can be isolated from menstrual blood. Mononuclear cells collected from the menstrual blood contained a subpopulation of adherent cells which could be maintained in tissue culture for >68 doublings and retained expression of the markers CD9, CD29, CD41a, CD44, CD59, CD73, CD90 and CD105, without karyotypic abnormalities. Proliferative rate of the cells was significantly higher than control umbilical cord derived mesenchymal stem cells, with doubling occurring every 19.4 hours. These cells, which we termed Endometrial Regenerative Cells (ERC) were capable of differentiating into 9 lineages: cardiomyocytic, respiratory epithelial, neurocytic, myocytic, endothelial, pancreatic, hepatic, adipocytic, and osteogenic. Additionally, ERC produced MMP3, MMP10, GM-CSF, angiopoietin-2 and PDGF-BB at 10–100,000 fold higher levels than two control cord blood derived mesenchymal stem cell lines. Given the ease of extraction and pluripotency of this cell population, we propose ERC as a novel alternative to current stem cells sources.


Journal of Translational Medicine | 2008

Allogeneic endometrial regenerative cells: An "Off the shelf solution" for critical limb ischemia?

Michael P. Murphy; Hao Wang; Amit N. Patel; Suman Kambhampati; Niren Angle; Kyle Chan; Annette M. Marleau; Andrew Pyszniak; Ewa Carrier; Thomas E. Ichim; Neil H. Riordan

Critical limb ischemia (CLI) is an advanced form of peripheral artery disease which is responsible for approximately 100,000 amputations per year in the US. Trials to date have reported clinical improvement and reduced need for amputation in CLI patients receiving autologous bone marrow or mobilized peripheral blood stem cells for stimulation of angiogenesis. While such treatments are currently entering Phase III trials, practical and scientific pitfalls will limit widespread implementation if efficacy is proven. Hurdles to be overcome include: a) reduced angiogenic potential of autologous cells in aged patients with cardiovascular risk factors; b) invasiveness/adverse effects of bone marrow extraction and G-CSF mobilization, respectively; and c) need for on-site cellular manipulation. The Endometrial Regenerative Cell (ERC) is a mesenchymal-like stem cell derived from the menstrual blood that is believed to be associated with endometrial angiogenesis. We discuss the possibility of using allogeneic ERCs as an off the shelf treatment for CLI based on the following properties: a) High levels of growth factors and matrix metalloprotease production; b) Ability to inhibits inflammatory responses and lack of immunogenicity; and c) Expandability to great quantities without loss of differentiation ability or karyotypic abnormalities.


Journal of Translational Medicine | 2009

Feasibility investigation of allogeneic endometrial regenerative cells.

Zhaohui Zhong; Amit N. Patel; Thomas E. Ichim; Neil H. Riordan; Hao Wang; Wei Ping Min; Erik J Woods; Michael A. Reid; Eduardo Mansilla; Gustavo Horacio Marín; Hugo Drago; Michael P. Murphy; Boris Minev

Endometrial Regenerative Cells (ERC) are a population of mesenchymal-like stem cells having pluripotent differentiation activity and ability to induce neoangiogenesis. In vitro and animal studies suggest ERC are immune privileged and in certain situations actively suppress ongoing immune responses. In this paper we describe the production of clinical grade ERC and initial safety experiences in 4 patients with multiple sclerosis treated intravenously and intrathecally. The case with the longest follow up, of more than one year, revealed no immunological reactions or treatment associated adverse effects. These preliminary data suggest feasibility of clinical ERC administration and support further studies with this novel stem cell type.


Nature Medicine | 2000

The axis of interleukin 12 and gamma interferon regulates acute vascularxenogeneic rejection

Hao Wang; Mark E. DeVries; Shaoping Deng; Masud H. Khandaker; J. Geoff Pickering; Larry H. Chow; Bertha Garcia; David J. Kelvin; Robert Zhong

Recent advances using transgenic animals or exogenous complement inhibitors have demonstrated prevention of hyperacute rejection of vascularized organs, but not graft loss due to acute vascular rejection. Using various wild-type and cytokine-deficient mice strains, we have examined the mechanisms of acute vascular rejection. C57BL/6 mice deficient in interleukin12 or gamma interferon showed faster acute vascular rejection than did wild-type mice. Furthermore, mice defective in B-cell development showed no acute vascular rejection. These results demonstrate that the axis of interleukin 12 and gamma interferon provides a survival advantage in vascularized xenografts by delaying or preventing acute vascular rejection caused by a B cell-dependent mechanism.


Kidney International | 2011

Carbon monoxide-releasing molecules protect against ischemia–reperfusion injury during kidney transplantation

Yves Caumartin; Jancy Stephen; Jian P. Deng; Dameng Lian; Zhu Lan; Weihua Liu; Bertha Garcia; Anthony M. Jevnikar; Hao Wang; Gediminas Cepinskas; Patrick Luke

Carbon monoxide (CO) can provide beneficial antiapoptotic and anti-inflammatory effects in the context of ischemia-reperfusion injury (IRI). Here we tested the ability of pretreating the kidney donor with carbon monoxide-releasing molecules (CORM) to prevent IRI in a transplant model. Isogeneic Brown Norway donor rats were pretreated with CORM-2 18u2009h before kidney retrieval. The kidneys were then cold-preserved for 26u2009h and transplanted into Lewis rat recipients that had undergone bilateral nephrectomy. Allografts from Brown Norway to Lewis rats were also performed after 6u2009h of cold ischemic time with low-dose tacrolimus treatment. All recipients receiving CORM-2-treated isografts survived the transplant process and had near-normal serum creatinine levels, whereas all control animals died of uremia by the third post-operative day. This beneficial effect was also seen in isografted Lewis recipients receiving kidneys perfused with CORM-3, indicating that CORMs have direct effects on the kidney. Pretreatment of human umbilical vein endothelial cells in culture with CORM-2 for 1u2009h significantly reduced cytokine-induced nicotinamide adenine dinucleotide phosphate-dependent production of superoxide, activation of the inflammation-relevant transcription factor nuclear factor-κB, upregulated expression of E-selectin and intercellular adhesion molecule-1 adhesion proteins, and leukocyte adhesion to the endothelial cells. Thus, CORM-2-derived CO protects renal transplants from IRI by modulating inflammation.


Transplantation | 1999

Complement inhibition with an anti-C5 monoclonal antibody prevents hyperacute rejection in a xenograft heart transplantation model

Hao Wang; Scott Rollins; Zuhua Gao; Bertha Garcia; Zheng Zhang; Jingjing Xing; Lan Li; Richard Kellersmann; Louis A. Matis; Robert Zhong

BACKGROUNDnThe present study was undertaken to determine whether anti-complement 5 (C5) monoclonal antibodies (mAb) prevent hyperacute rejection (HAR) in a rat-to-presensitized mouse heart transplantation model and whether these mAb, combined with cyclosporine (CsA) and cyclophosphamide (CyP), can achieve long-term graft survival.nnnMETHODSnBALB/c mice were presensitized with 2x10(7) splenocytes from Lewis rats 14 days before grafting. Heart grafts from Lewis rats were heterotopically transplanted into BALB/c mice. Presensitized mice were treated with either anti-C5 mAb or a combination of anti-C5 mAb, CsA, and CyP. Controls included: presensitized mice with no treatment, presensitized mice treated with either CsA + CyP or IgG, and nonpresensitized mice with either no treatment or with CsA + CyP treatment.nnnRESULTSnAlthough typical features of HAR were evident in the presensitized grafts, the mAb completely inhibited complement activation and successfully prevented HAR. Despite complement inactivation, the graft was rejected on postoperative day 6 with acute vascular rejection (AVR) also known as delayed xenograft rejection (DXR). Notably, this type of rejection cannot be effectively overcome by CsA and CyP.nnnCONCLUSIONSnWe conclude that (1) anti-C5 mAb prevents HAR, (2) AVR/DXR still occurs when HAR is prevented by complement inactivation, and (3) AVR/DXR cannot be overcome by conventional immunosuppression. These data suggest that anti-C5 mAb may be valuable for preventing HAR in future clinical xenotransplantation and that additional interventions may be required to address AVR/DXR.


Journal of Immunology | 2003

Cytokines Regulate the Pattern of Rejection and Susceptibility to Cyclosporine Therapy in Different Mouse Recipient Strains After Cardiac Allografting

Hao Wang; Karoline A. Hosiawa; Wei-Ping Min; Jinming Yang; Xiaoxia Zhang; Bertha Garcia; Thomas E. Ichim; Dejun Zhou; Dameng Lian; David J. Kelvin; Robert Zhong

We determined the role of cytokines in regulating the pattern of rejection and recipient susceptibility to cyclosporine (CsA) in a mouse cardiac allograft model. Hearts from C3H mice transplanted into untreated BALB/c (Th2-dominant) and C57BL/6 (Th1-dominant) mice showed different patterns of rejection. C3H allografts in BALB/c mice showed typical acute vascular rejection (AVR) with strong intragraft deposition and high serum levels of anti-donor IgG with predominant IgG1, while C3H allografts in C57BL/6 mice showed typical acute cellular rejection (ACR) with massive intragraft infiltration of CD4+ and CD8+ lymphocytes and low serum levels of anti-donor IgG with predominant IgG2a. Elevated intragraft mRNA expression of IL-2, IFN-γ, and IL-12 mRNA was present in C57BL/6 recipients, whereas allografts in BALB/c mice displayed increased IL-4 and IL-10 mRNA levels. CsA therapy completely inhibited ACR and induced indefinite allograft survival in C57BL/6 recipients, while the same therapy failed to prevent AVR, and only marginally prolonged graft survival in BALB/c recipients. In contrast, rapamycin blocked AVR, achieving indefinite survival in BALB/c recipients, but was less effective at preventing ACR in C57BL/6 recipients. The disruption of the IL-12 or IFN-γ genes in C57BL/6 mice shifted ACR to AVR, and resulted in concomitant recipient resistance to CsA therapy. Conversely, disruption of IL-4 gene in BALB/c mice markedly attenuated AVR and significantly prolonged allograft survival. These data suggest that the distinct cytokine profiles expressed by different mouse strains play an essential role in regulating the pattern of rejection and outcome of CsA/rapamycin therapy.


Seminars in Immunology | 2003

The role of chemokines and chemokine receptors in alloantigen-independent and alloantigen-dependent transplantation injury.

Mark E. DeVries; Karoline A. Hosiawa; Cheryl M. Cameron; Steven E. Bosinger; Desmond Persad; Alyson A. Kelvin; Jeff C. Coombs; Hao Wang; Robert Zhong; Mark J. Cameron; David J. Kelvin

Transplantation injury and rejection involves the interplay of innate and acquired immune responses. Immune-related injury manifests itself in three temporal phases: early innate immune driven alloantigen-independent injury, acquired immune driven alloantigen-dependent injury, and chronic injury. Sequential waves of chemokine expression play a central role in regulating graft injury through the recruitment of phagocytes shortly after transplantation and activated lymphocytes and phagocytes in the weeks and years following transplantation. This review focuses on recent studies demonstrating the role of chemokines in transplantation.


Transplant Immunology | 2008

Reduction of Foxp3-expressing regulatory T cell infiltrates during the progression of renal allograft rejection in a mouse model

Shuang Wang; Jifu Jiang; Qiunong Guan; Zhu Lan; Hao Wang; Christopher Y. Nguan; Anthony M. Jevnikar; Caigan Du

BACKGROUNDnRegulatory T (Treg) cells are the immune suppressors in the maintenance of immune homeostasis and tolerance to self and non-self antigens, and may have therapeutic potential in the treatment of transplant rejection in patients. However, Treg cell development and action are poorly understood in transplantation. In this study, the association of CD4(+)Foxp3(+) infiltrates within renal allograft tissue with graft survival was investigated in a mouse model.nnnMETHODSnKidney donors from C57BL/6J mice (H-2(b)) were transplanted to bilaterally nephrectomized Balb/c recipient mice (H-2(d)). Treg cells were examined with FACS and immunohistochemical staining.nnnRESULTSnHere we showed that without any immunosuppressive regimen, kidney allografts were mostly rejected from 20 to 60 days after transplantation. During the progression of allograft rejection Foxp3(+) Treg phenotype infiltrates were significantly diminished, while intragraft expression of TGF-beta1, IL-6, IL-17 and IL-23 was up-regulated. The regulatory function of CD4(+)CD25(+) infiltrates was confirmed by their suppressive activity in mixed lymphocyte reaction. Further in vitro studies indicated that primary renal tubular epithelial cell (TEC) cultures produced high levels of IL-6 in response to allogeneic lymphocyte or IL-17 stimulation, and neutralization of IL-6 increased CD4(+)CD25(+)Foxp3(+) cells in co-cultures with TEC.nnnCONCLUSIONnDiminution of Foxp3(+) Treg infiltrates associates with renal allograft rejection, and neutralization of IL-6 activity enhances Foxp3(+) Treg cell differentiation. Our findings suggest that increase in intragraft IL-6 may down-regulate infiltrating Foxp3(+) Treg cells.


Xenotransplantation | 2003

Treatment with a short course of LF 15-0195 and continuous cyclosporin A attenuates acute xenograft rejection in a rat-to-mouse cardiac transplantation model.

Hao Wang; Karoline A. Hosiawa; Bertha Garcia; Jeffrey Shum; Patrick Dutartre; David J. Kelvin; Robert Zhong

Abstract:u2002Searching for a novel immunosuppressive agent to effectively prevent acute vascular rejection (AVR) is essential for success in clinical xenotransplantation. We previously reported that Lewis rat hearts transplanted into BALB/c mice developed typical AVR in 6u2003days. The present study was undertaken to determine the efficacy of LF 15‐0195, a new immunosuppressive analog of 15‐deoxyspergualin in the prevention of AVR in a rat‐to‐mouse cardiac xenograft model. We transplanted 2‐week old Lewis rat hearts into BALB/c mice. Four groups were included in this study: untreated recipients and cyclosporin A (CsA) treated recipients were controls; LF 15‐0195 treated recipients or LF 15‐0195 combined with CsA treated recipients were experimental groups. Mouse recipients received either LF 15‐0195 2u2003mg/kg subcutaneously from day‐1 to post‐operative day 14, or CsA 15u2003mg/kg subcutaneously daily, from day 0 to endpoint rejection, or the two drugs in combination. We observed that high dose CsA did not inhibit AVR and the graft was rejected in 11.3u2003±u20031.9u2003days. Graft histology and immunohistology showed typical AVR, characterized by interstitial hemorrhage, intravascular fibrin deposition, thrombosis, and massive deposition of anti‐rat immunoglobulin G (IgG) and immunoglobulin M (IgM). Serum xenoreactive antibodies (xAbs) were markedly elevated in these animals as well. In contrast, we observed that treatment with LF 15‐0195 alone significantly prolonged graft survival to 19.3u2003±u20030.7u2003days. Notably, xAbs were significantly decreased and the rejection pattern of these grafts was cell‐mediated rejection (CMR), instead of AVR. When CsA was combined with LF 15‐0195, the graft mean survival time was further increased to 58.5u2003±u200317.3u2003days. Antibody production and T‐cell infiltration were significantly inhibited at the terminal stages of graft survival and pathology showed striking attenuation of both AVR and CMR. Sequential studies on days 6 and 14 demonstrated that LF 15‐0195 either alone or combined with CsA completely inhibited antibody production. However, intragraft infiltration by Mac‐1 positive cells including natural killer cells, macrophages and granulocytes in LF 15‐0195 treated recipients was similar to that of untreated recipients. We conclude that LF 15‐0195 effectively prevented AVR by markedly inhibiting the production of anti‐donor IgG xAbs. Also, treatment with short course LF 15‐0195 and continuous CsA significantly reduced T‐cell infiltration. Studies to test this therapy in inhibiting AVR in a pig‐to‐non‐human primate xenotransplantation model are underway.

Collaboration


Dive into the Hao Wang's collaboration.

Top Co-Authors

Avatar

Bertha Garcia

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Robert Zhong

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Jacqueline Arp

Robarts Research Institute

View shared research outputs
Top Co-Authors

Avatar

Weihua Liu

Lawson Health Research Institute

View shared research outputs
Top Co-Authors

Avatar

Anthony M. Jevnikar

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Jifu Jiang

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

David J. Kelvin

University Health Network

View shared research outputs
Top Co-Authors

Avatar

Siobhan Ramcharran

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Wei Ge

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Zhu Lan

University of Western Ontario

View shared research outputs
Researchain Logo
Decentralizing Knowledge