1
|
Li B, Djahanpour N, Zamzam A, Syed MH, Jain S, Abdin R, Qadura M. Angiogenesis-related proteins as biomarkers for peripheral artery disease. Heliyon 2023; 9:e20166. [PMID: 37809892 PMCID: PMC10559913 DOI: 10.1016/j.heliyon.2023.e20166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 09/07/2023] [Accepted: 09/13/2023] [Indexed: 10/10/2023] Open
Abstract
Background Angiogenesis plays an important role in peripheral artery disease (PAD) and angiogenesis-related proteins may act as prognostic biomarkers. This study assesses the potential for angiogenesis-related proteins to predict adverse events associated with PAD. Methods This was a case-control study. Patients with PAD (n = 250) and without PAD (n = 125) provided blood samples and were followed prospectively for three years. Concentrations of 17 angiogenesis-related proteins were measured in plasma. The incidence of major adverse limb event (MALE), defined as a composite of major amputation or vascular intervention, was the primary outcome. Worsening PAD status, defined as a drop in ankle brachial index ≥ 0.15, was the secondary outcome. Multivariable regression adjusted for baseline characteristics was conducted to determine the prognostication value of angiogenesis-related proteins in predicting MALE. Findings Relative to patients without PAD, 8 proteins related to angiogenesis were expressed differentially in PAD patients. Worsening PAD status and MALE were observed in 52 (14%) and 83 (22%) patients, respectively. Hepatocyte growth factor (HGF) was the most reliable predictor of MALE (adjusted HR 0.79, 95% CI 0.15-0.86). Compared to individuals with high HGF, patients with low HGF had a decreased three-year freedom from MALE [66% vs 88%, p = 0.001], major amputation [93% vs 98%, p = 0.023], vascular intervention [68% vs 88%, p = 0.001], and worsening PAD status [81% vs 91%, p = 0.006]. Interpretation Measuring plasma levels of HGF in individuals with PAD can assist in identifying patients at elevated risk of adverse events related to PAD who may benefit from additional evaluation or treatment.
Collapse
Affiliation(s)
- Ben Li
- Division of Vascular Surgery, St. Michael's Hospital, Unity Health Toronto, University of Toronto, Canada
| | - Niousha Djahanpour
- Division of Vascular Surgery, St. Michael's Hospital, Unity Health Toronto, University of Toronto, Canada
| | - Abdelrahman Zamzam
- Division of Vascular Surgery, St. Michael's Hospital, Unity Health Toronto, University of Toronto, Canada
| | - Muzammil H. Syed
- Division of Vascular Surgery, St. Michael's Hospital, Unity Health Toronto, University of Toronto, Canada
| | - Shubha Jain
- Division of Vascular Surgery, St. Michael's Hospital, Unity Health Toronto, University of Toronto, Canada
| | - Rawand Abdin
- Department of Medicine, McMaster University, Hamilton, Canada
| | - Mohammad Qadura
- Division of Vascular Surgery, St. Michael's Hospital, Unity Health Toronto, University of Toronto, Canada
- Department of Surgery, University of Toronto, Toronto, Canada
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Unity Health Toronto, University of Toronto, Canada
| |
Collapse
|
2
|
Palma Medina LM, Babačić H, Dzidic M, Parke Å, Garcia M, Maleki KT, Unge C, Lourda M, Kvedaraite E, Chen P, Muvva JR, Cornillet M, Emgård J, Moll K, Michaëlsson J, Flodström-Tullberg M, Brighenti S, Buggert M, Mjösberg J, Malmberg KJ, Sandberg JK, Gredmark-Russ S, Rooyackers O, Svensson M, Chambers BJ, Eriksson LI, Pernemalm M, Björkström NK, Aleman S, Ljunggren HG, Klingström J, Strålin K, Norrby-Teglund A. Targeted plasma proteomics reveals signatures discriminating COVID-19 from sepsis with pneumonia. Respir Res 2023; 24:62. [PMID: 36829233 PMCID: PMC9950694 DOI: 10.1186/s12931-023-02364-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 02/10/2023] [Indexed: 02/26/2023] Open
Abstract
BACKGROUND COVID-19 remains a major public health challenge, requiring the development of tools to improve diagnosis and inform therapeutic decisions. As dysregulated inflammation and coagulation responses have been implicated in the pathophysiology of COVID-19 and sepsis, we studied their plasma proteome profiles to delineate similarities from specific features. METHODS We measured 276 plasma proteins involved in Inflammation, organ damage, immune response and coagulation in healthy controls, COVID-19 patients during acute and convalescence phase, and sepsis patients; the latter included (i) community-acquired pneumonia (CAP) caused by Influenza, (ii) bacterial CAP, (iii) non-pneumonia sepsis, and (iv) septic shock patients. RESULTS We identified a core response to infection consisting of 42 proteins altered in both COVID-19 and sepsis, although higher levels of cytokine storm-associated proteins were evident in sepsis. Furthermore, microbiologic etiology and clinical endotypes were linked to unique signatures. Finally, through machine learning, we identified biomarkers, such as TRIM21, PTN and CASP8, that accurately differentiated COVID-19 from CAP-sepsis with higher accuracy than standard clinical markers. CONCLUSIONS This study extends the understanding of host responses underlying sepsis and COVID-19, indicating varying disease mechanisms with unique signatures. These diagnostic and severity signatures are candidates for the development of personalized management of COVID-19 and sepsis.
Collapse
Affiliation(s)
- Laura M. Palma Medina
- grid.24381.3c0000 0000 9241 5705Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 141 52, Stockholm, Sweden
| | - Haris Babačić
- grid.4714.60000 0004 1937 0626Science for Life Laboratory, Department of Oncology and Pathology, Karolinska Institute, Stockholm, Sweden
| | - Majda Dzidic
- grid.24381.3c0000 0000 9241 5705Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 141 52, Stockholm, Sweden
| | - Åsa Parke
- grid.4714.60000 0004 1937 0626Department of Medicine Huddinge, Karolinska Institute, Stockholm, Sweden ,grid.24381.3c0000 0000 9241 5705Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Marina Garcia
- grid.24381.3c0000 0000 9241 5705Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 141 52, Stockholm, Sweden
| | - Kimia T. Maleki
- grid.24381.3c0000 0000 9241 5705Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 141 52, Stockholm, Sweden
| | - Christian Unge
- grid.4714.60000 0004 1937 0626Department of Medicine Huddinge, Karolinska Institute, Stockholm, Sweden ,grid.24381.3c0000 0000 9241 5705Functional Area of Emergency Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Magda Lourda
- grid.24381.3c0000 0000 9241 5705Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 141 52, Stockholm, Sweden ,grid.4714.60000 0004 1937 0626Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| | - Egle Kvedaraite
- grid.24381.3c0000 0000 9241 5705Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 141 52, Stockholm, Sweden ,grid.24381.3c0000 0000 9241 5705Department of Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
| | - Puran Chen
- grid.24381.3c0000 0000 9241 5705Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 141 52, Stockholm, Sweden
| | - Jagadeeswara Rao Muvva
- grid.24381.3c0000 0000 9241 5705Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 141 52, Stockholm, Sweden
| | - Martin Cornillet
- grid.24381.3c0000 0000 9241 5705Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 141 52, Stockholm, Sweden
| | - Johanna Emgård
- grid.24381.3c0000 0000 9241 5705Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 141 52, Stockholm, Sweden
| | - Kirsten Moll
- grid.24381.3c0000 0000 9241 5705Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 141 52, Stockholm, Sweden
| | | | - Jakob Michaëlsson
- grid.24381.3c0000 0000 9241 5705Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 141 52, Stockholm, Sweden
| | - Malin Flodström-Tullberg
- grid.24381.3c0000 0000 9241 5705Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 141 52, Stockholm, Sweden
| | - Susanna Brighenti
- grid.24381.3c0000 0000 9241 5705Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 141 52, Stockholm, Sweden
| | - Marcus Buggert
- grid.24381.3c0000 0000 9241 5705Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 141 52, Stockholm, Sweden
| | - Jenny Mjösberg
- grid.24381.3c0000 0000 9241 5705Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 141 52, Stockholm, Sweden
| | - Karl-Johan Malmberg
- grid.24381.3c0000 0000 9241 5705Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 141 52, Stockholm, Sweden
| | - Johan K. Sandberg
- grid.24381.3c0000 0000 9241 5705Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 141 52, Stockholm, Sweden
| | - Sara Gredmark-Russ
- grid.24381.3c0000 0000 9241 5705Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 141 52, Stockholm, Sweden ,grid.24381.3c0000 0000 9241 5705Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden ,The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå, Sweden
| | - Olav Rooyackers
- grid.24381.3c0000 0000 9241 5705Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden ,grid.4714.60000 0004 1937 0626Division for Anesthesiology and Intensive Care, Department of Clinical Interventions and Technology CLINTEC, Karolinska Institutet, Stockholm, Sweden
| | - Mattias Svensson
- grid.24381.3c0000 0000 9241 5705Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 141 52, Stockholm, Sweden
| | - Benedict J. Chambers
- grid.24381.3c0000 0000 9241 5705Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 141 52, Stockholm, Sweden
| | - Lars I. Eriksson
- grid.24381.3c0000 0000 9241 5705Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden
| | - Maria Pernemalm
- grid.4714.60000 0004 1937 0626Science for Life Laboratory, Department of Oncology and Pathology, Karolinska Institute, Stockholm, Sweden
| | - Niklas K. Björkström
- grid.24381.3c0000 0000 9241 5705Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 141 52, Stockholm, Sweden
| | - Soo Aleman
- grid.4714.60000 0004 1937 0626Department of Medicine Huddinge, Karolinska Institute, Stockholm, Sweden ,grid.24381.3c0000 0000 9241 5705Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Hans-Gustaf Ljunggren
- grid.24381.3c0000 0000 9241 5705Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 141 52, Stockholm, Sweden
| | - Jonas Klingström
- grid.24381.3c0000 0000 9241 5705Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 141 52, Stockholm, Sweden
| | - Kristoffer Strålin
- grid.4714.60000 0004 1937 0626Department of Medicine Huddinge, Karolinska Institute, Stockholm, Sweden ,grid.24381.3c0000 0000 9241 5705Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Norrby-Teglund
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 141 52, Stockholm, Sweden.
| |
Collapse
|
3
|
Yun JH. Hepatocyte growth factor prevents pericyte loss in diabetic retinopathy. Microvasc Res 2021; 133:104103. [PMID: 33181170 DOI: 10.1016/j.mvr.2020.104103] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/26/2020] [Accepted: 11/04/2020] [Indexed: 01/21/2023]
Abstract
Diabetic retinopathy (DR) is a disease that causes blindness due to vascular leakage or abnormal angiogenesis. Hepatocyte growth factor (HGF) is increased in the serum or vitreous fluid in proliferative diabetic retinopathy (PDR) patients, although the effect of HGF on the blood vessels remains unclear. This study focused on the effect of HGF on pericyte (PC) survival and endothelial cell (EC) permeability. It was demonstrated that HGF was increased in the diabetic mouse retina. However, HGF prevented PC apoptosis caused by TNF-α, which increased in the diabetic retinas both in vitro and in vivo. In addition, HGF was involved in PC survival by increasing the Akt signaling pathway. Moreover, HGF strengthened the EC tight junction in co-cultures of PCs and ECs by promoting PC survival, thereby reducing EC permeability. These results suggest that HGF may play a role in the prevention of increased vascular leakage by inhibiting the PC loss that occurs in DR to some extent. However, careful HGF reduction in DR might avoid an increase in PC loss.
Collapse
Affiliation(s)
- Jang-Hyuk Yun
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Gangwon, Republic of Korea; College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, 1 Gangwondaehakgil, Chuncheon-si, Gangwon-do 24341, Republic of Korea.
| |
Collapse
|
4
|
Akolpoglu MB, Inceoglu Y, Bozuyuk U, Sousa AR, Oliveira MB, Mano JF, Kizilel S. Recent advances in the design of implantable insulin secreting heterocellular islet organoids. Biomaterials 2020; 269:120627. [PMID: 33401104 DOI: 10.1016/j.biomaterials.2020.120627] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 12/14/2020] [Accepted: 12/18/2020] [Indexed: 12/11/2022]
Abstract
Islet transplantation has proved one of the most remarkable transmissions from an experimental curiosity into a routine clinical application for the treatment of type I diabetes (T1D). Current efforts for taking this technology one-step further are now focusing on overcoming islet donor shortage, engraftment, prolonged islet availability, post-transplant vascularization, and coming up with new strategies to eliminate lifelong immunosuppression. To this end, insulin secreting 3D cell clusters composed of different types of cells, also referred as heterocellular islet organoids, spheroids, or pseudoislets, have been engineered to overcome the challenges encountered by the current islet transplantation protocols. β-cells or native islets are accompanied by helper cells, also referred to as accessory cells, to generate a cell cluster that is not only able to accurately secrete insulin in response to glucose, but also superior in terms of other key features (e.g. maintaining a vasculature, longer durability in vivo and not necessitating immunosuppression after transplantation). Over the past decade, numerous 3D cell culture techniques have been integrated to create an engineered heterocellular islet organoid that addresses current obstacles. Here, we first discuss the different cell types used to prepare heterocellular organoids for islet transplantation and their contribution to the organoids design. We then introduce various cell culture techniques that are incorporated to prepare a fully functional and insulin secreting organoids with select features. Finally, we discuss the challenges and present a future outlook for improving clinical outcomes of islet transplantation.
Collapse
Affiliation(s)
- M Birgul Akolpoglu
- Chemical and Biological Engineering, Koc University, Sariyer, 34450, Istanbul, Turkey
| | - Yasemin Inceoglu
- Chemical and Biological Engineering, Koc University, Sariyer, 34450, Istanbul, Turkey
| | - Ugur Bozuyuk
- Chemical and Biological Engineering, Koc University, Sariyer, 34450, Istanbul, Turkey
| | - Ana Rita Sousa
- Department of Chemistry, CICECO - Aveiro Institute of Materials. University of Aveiro. Campus Universitário de Santiago. 3810-193 Aveiro. Portugal
| | - Mariana B Oliveira
- Department of Chemistry, CICECO - Aveiro Institute of Materials. University of Aveiro. Campus Universitário de Santiago. 3810-193 Aveiro. Portugal.
| | - João F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials. University of Aveiro. Campus Universitário de Santiago. 3810-193 Aveiro. Portugal
| | - Seda Kizilel
- Chemical and Biological Engineering, Koc University, Sariyer, 34450, Istanbul, Turkey.
| |
Collapse
|
5
|
Reyes Valenzuela A, Bao G, Vikstrom A, Kost KM, Prakash S, Mongeau L. Polymeric Microspheres Containing Human Vocal Fold Fibroblasts for Vocal Fold Regeneration. Laryngoscope 2020; 131:1828-1834. [PMID: 33068297 DOI: 10.1002/lary.29118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 08/07/2020] [Accepted: 09/08/2020] [Indexed: 01/27/2023]
Abstract
OBJECTIVE Most acellular injectable biomaterials for vocal fold (VF) wound treatment have limited regenerative potential due to their fast enzymatic degradation and limited recruitment of native cells postinjection. The injection of cells as therapeutic treatment often results in apoptosis due to stresses within the needle and the immune response of the host. Degradable microspheres may improve treatment effectiveness by increasing cell residence time, shielding cells during injection, and offering early protection against the immune system response. The objective of the present study was to investigate the potential of human VF fibroblasts encapsulated in polymeric microspheres as an injectable therapeutic treatment in vitro. METHODS Alginate, alginate-poly-L-lysine, and alginate-chitosan microspheres were fabricated using electrospraying and characterized in terms of biocompatibility, swelling, and mechanical properties as well as cytokine production. RESULTS Alginate microspheres were found to have the most desirable properties for VF regeneration. They were resistant to mechanical challenges. They were found to have a stiffness similar to that reported for native VF-lamina propria. They were found to be biocompatible and increased the proliferation of fibroblasts. Human VF fibroblasts encapsulated in alginate microspheres induced the production of interleukin (IL)-8 and IL-4 at 24 hours. CONCLUSION The alginate microspheres fabricated in this study were found to offer potential advantages, as cell delivery tool. This study highlights the importance of combining biomaterials and cells to expedite the wound-healing process through cytokine production. Future work is aimed to further analysis of the wound-healing properties the microspheres. LEVEL OF EVIDENCE NA Laryngoscope, 131:1828-1834, 2021.
Collapse
Affiliation(s)
- Alicia Reyes Valenzuela
- Biomedical and Cell Therapy Research Laboratory, Department of Biomedical Engineering and Artificial Cells and Organs Research Center, Faculty of Medicine, McGill University, Montreal, Quebec, Canada.,Department of Mechanical Engineering, Faculty of Engineering, McGill University, Montreal, Quebec, Canada
| | - Guangyu Bao
- Department of Mechanical Engineering, Faculty of Engineering, McGill University, Montreal, Quebec, Canada
| | - Abigail Vikstrom
- Department of Mechanical Engineering, Faculty of Engineering, McGill University, Montreal, Quebec, Canada
| | - Karen M Kost
- Department of Otolaryngology - Head and Neck Surgery, McGill University Voice and Dysphagia Laboratory, Montreal, Quebec, Canada
| | - Satya Prakash
- Biomedical and Cell Therapy Research Laboratory, Department of Biomedical Engineering and Artificial Cells and Organs Research Center, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Luc Mongeau
- Department of Mechanical Engineering, Faculty of Engineering, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
6
|
Coon JI, Jain S, Sepuru KM, Chung Y, Mohankumar K, Rajarathnam K, Jain SK. Lyophilization of human amniotic fluid is feasible without affecting biological activity. Pediatr Res 2020; 87:847-852. [PMID: 31756731 DOI: 10.1038/s41390-019-0632-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 09/20/2019] [Accepted: 10/02/2019] [Indexed: 11/09/2022]
Abstract
BACKGROUND Fetal swallowing of human amniotic fluid (hAF) containing trophic factors (TFs) promotes gastrointestinal tract (GIT) development. Preterm birth interrupts hAF swallowing, which may increase the risk of necrotizing enterocolitis (NEC). Postnatally, it is difficult to replicate fetal swallowing of hAF due to volume. We aimed to evaluate whether hAF lyophilization is feasible and its effect on hAF-borne TFs. METHODS We collected hAF (n = 16) from uncomplicated pregnancies. hAF was divided into three groups: unprocessed control (C), concentration by microfiltration (F), and by dialysis and lyophilization (L). EGF, HGF, GM-CSF, and TGF-α were measured in each group by multiplex assay. Bioavailability of TFs was measured by proliferation and LPS-induced IL-8 production by intestinal epithelial cells FHs74. RESULTS After dialysis/lyophilization, GM-CSF and TGF-α were preserved with partial loss of EGF and HGF. hAF increased cell proliferation and reduced LPS-induced IL-8 production compared to medium alone. Compared to control, dialysis/lyophilization and filtration of hAF increased FHs74 cell proliferation (p < 0.001) and decreased LPS-induced IL-8 production (p < 0.01). CONCLUSIONS Lyophilization and filtration of hAF is feasible with partial loss of TFs but maintains and even improves bioavailability of TFs measured by proliferation and LPS-induced IL-8 production by FHs74.
Collapse
Affiliation(s)
- John I Coon
- Department of Pediatrics, Division of Neonatology, University of Texas Medical Branch, Galveston, TX, USA
| | - Sangeeta Jain
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - Krishna M Sepuru
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Yerin Chung
- Department of Pediatrics, Division of Neonatology, John Hopkins University, Baltimore, MD, USA
| | - Krishnan Mohankumar
- Department of Pediatrics, Division of Neonatology, John Hopkins University, Baltimore, MD, USA
| | - Krishna Rajarathnam
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Sunil K Jain
- Department of Pediatrics, Division of Neonatology, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
7
|
MET targeting: time for a rematch. Oncogene 2020; 39:2845-2862. [PMID: 32034310 DOI: 10.1038/s41388-020-1193-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/16/2020] [Accepted: 01/24/2020] [Indexed: 12/21/2022]
Abstract
MET, the receptor tyrosine kinase (RTK) for hepatocyte growth factor, is a proto-oncogene involved in embryonic development and throughout life in homeostasis and tissue regeneration. Deregulation of MET signaling has been reported in numerous malignancies, prompting great interest in MET targeting for cancer therapy. The present review offers a summary of the biology of MET and its known functions in normal physiology and carcinogenesis, followed by an overview of the most relevant MET-targeting strategies and corresponding clinical trials, highlighting both past setbacks and promising future prospects. By placing their efforts on a more precise stratification strategy through the genetic analysis of tumors, modern trials such as the NCI-MATCH trial could revive the past enthusiasm for MET-targeted therapy.
Collapse
|
8
|
Wang L, Zhao Y, Yang F, Feng M, Zhao Y, Chen X, Mi J, Yao Y, Guan D, Xiao Z, Chen B, Dai J. Biomimetic collagen biomaterial induces in situ lung regeneration by forming functional alveolar. Biomaterials 2020; 236:119825. [PMID: 32044576 DOI: 10.1016/j.biomaterials.2020.119825] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 01/07/2020] [Accepted: 01/25/2020] [Indexed: 01/02/2023]
Abstract
In situ restoration of severely damaged lung remains difficult due to its limited regeneration capacity after injury. Artificial lung scaffolds are emerging as potential substitutes, but it is still a challenge to reconstruct lung regeneration microenvironment in scaffold after lung resection injury. Here, a 3D biomimetic porous collagen scaffold with similar structure characteristics as lung is fabricated, and a novel collagen binding hepatocyte growth factor (CBD-HGF) is tethered on the collagen scaffold for maintaining the biomimetic function of HGF to improve the lung regeneration microenvironment. The biomimetic scaffold was implanted into the operative region of a rat partial lung resection model. The results revealed that vascular endothelial cells and endogenous alveolar stem cells entered the scaffold at the early stage of regeneration. At the later stage, inflammation and fibrosis were attenuated, the microvascular and functional alveolar-like structures were formed, and the general morphology of the injured lung was restored. Taken together, the functional 3D biomimetic collagen scaffold facilitates recovery of the injured lung, alveolar regeneration, and angiogenesis after acute lung injury. Particularly, this is the first study of lung regeneration in vivo guided by biomimetic collagen scaffold materials, which supports the concept that tissue engineering is an effective strategy for alveolar regeneration.
Collapse
Affiliation(s)
- Linjie Wang
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Yannan Zhao
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China; State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Feng Yang
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Meng Feng
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Yazhen Zhao
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Xi Chen
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Junwei Mi
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Yuanjiang Yao
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Dongwei Guan
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Zhifeng Xiao
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China; State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Bing Chen
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China; State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Jianwu Dai
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China; State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
9
|
Gao X, Jiang P, Zhang Q, Liu Q, Jiang S, Liu L, Guo M, Cheng Q, Zheng J, Yao H. Peglated-H1/pHGFK1 nanoparticles enhance anti-tumor effects of sorafenib by inhibition of drug-induced autophagy and stemness in renal cell carcinoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:362. [PMID: 31426831 PMCID: PMC6699135 DOI: 10.1186/s13046-019-1348-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 07/29/2019] [Indexed: 12/19/2022]
Abstract
Background Tumor targeting small molecular inhibitors are the most popular treatments for many malignant diseases, including cancer. However, the lower clinical response and drug resistance still limit their clinical efficacies. HGFK1, the first kringle domain of hepatocyte growth factor, has been defined as a potent anti-angiogenic factor. Here, we aimed to develop and identify novel nanoparticles—PH1/pHGFK1 as potential therapeutic agents for the treatment of renal cell carcinoma (RCC). Methods We produced a novel cationic polymer—PH1 and investigated the anti-tumor activity of PH1/pHGFK1 nanoparticle alone and its combination therapy with sorafenib in RCC cell line xenografted mice model. Then, we figured out its molecular mechanisms in human RCC cell lines in vitro. Results We firstly demonstrated that intravenous injection of PH1/pHGFK1 nanoparticles significantly inhibited tumor growth and prolonged the survival time of tumor-bearing mice, as well as synergistically enhanced anti-tumor activities of sorafenib. Furthermore, we elucidated that recombinant HGFK1 improved sorafenib-induced cell apoptosis and arrested cell cycle. In addition, HGFK1 could also decrease sorafenib-induced autophagy and stemness via blockading NF-κB signaling pathway in RCC both in vitro and in vivo. Conclusions HGFK1 could inhibit tumor growth, synergistically enhance anti-tumor activities of sorafenib and reverse its drug resistance evolution in RCC. Our results provide rational basis for clinical application of sorafenib and HGFK1 combination therapy in RCC patients. Electronic supplementary material The online version of this article (10.1186/s13046-019-1348-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaoge Gao
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province, 221002, People's Republic of China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, 221002, People's Republic of China
| | - Pin Jiang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province, 221002, People's Republic of China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, 221002, People's Republic of China
| | - Qian Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province, 221002, People's Republic of China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, 221002, People's Republic of China
| | - Qian Liu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province, 221002, People's Republic of China
| | - Shuangshuang Jiang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province, 221002, People's Republic of China
| | - Ling Liu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province, 221002, People's Republic of China
| | - Maomao Guo
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province, 221002, People's Republic of China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, 221002, People's Republic of China
| | - Qian Cheng
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province, 221002, People's Republic of China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, 221002, People's Republic of China
| | - Junnian Zheng
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province, 221002, People's Republic of China. .,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, 221002, People's Republic of China.
| | - Hong Yao
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province, 221002, People's Republic of China. .,Department of Cancer Biotherapy Center, Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650118, People's Republic of China.
| |
Collapse
|
10
|
In vitro differentiation of human umbilical cord blood mesenchymal stem cells into functioning hepatocytes. ALEXANDRIA JOURNAL OF MEDICINE 2019. [DOI: 10.1016/j.ajme.2016.05.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
11
|
A Sputum Screening Test to Rule Out Pneumonia at an Early Stage With High Negative Predictive Value. POINT OF CARE 2018. [DOI: 10.1097/poc.0000000000000170] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
12
|
Bellan M, Castello LM, Pirisi M. Candidate Biomarkers of Liver Fibrosis: A Concise, Pathophysiology-oriented Review. J Clin Transl Hepatol 2018; 6:317-325. [PMID: 30271745 PMCID: PMC6160308 DOI: 10.14218/jcth.2018.00006] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 04/25/2018] [Accepted: 05/03/2018] [Indexed: 12/11/2022] Open
Abstract
Repair of sustained liver injury results in fibrosis (i.e. the accumulation of extracellular matrix proteins), and ultimately the complete distortion of parenchymal architecture of the liver, which we call cirrhosis. Detecting and staging of fibrosis is thus a mainstay in the management of chronic liver diseases, since many clinically relevant decisions, such as starting treatment and/or monitoring for complications including hepatocellular carcinoma, may depend on it. The gold standard for fibrosis staging is liver biopsy, the role of which, however, is questioned nowadays because of cost, hazards and poor acceptance by patients. On the other hand, imaging techniques and/or measurement of direct and indirect serum markers have not proved to be completely satisfactory under all circumstances as alternatives to liver biopsy. Making progress in this field is now more crucial than ever, since treatments for established fibrosis appear on the horizon. Fine dissection of the pathways involved in the pathophysiology of liver diseases has put forward several novel candidate biomarkers of liver fibrosis, such as growth arrest-specific6, Mac-2-binding protein, osteopontin, placental growth factor, growth/differentiation factor 15 and hepatocyte growth factor. All molecules have been suggested to have potential to complement or substitute methods currently used to stage liver diseases. Here, we review the pros and cons for their use in this setting.
Collapse
Affiliation(s)
- Mattia Bellan
- Department of Translational Medicine, Università del Piemonte Orientale UPO, Novara, Italy
- Division of Internal Medicine, “Sant’Andrea Hospital”, Vercelli, Italy
- IRCAD, Interdisciplinary Research Center of Autoimmune Diseases, Novara, Italy
- *Correspondence to: Mattia Bellan, Department of Translational Medicine, Università del Piemonte Orientale UPO, via Solaroli 17, Novara (NO) 28100, Italy. Tel: +39-321-3733966, Fax: +39-321-3733361, E-mail:
| | - Luigi Mario Castello
- Department of Translational Medicine, Università del Piemonte Orientale UPO, Novara, Italy
- Emergency Medicine Department, “AOU Maggiore della Carità”, Novara, Italy
| | - Mario Pirisi
- Department of Translational Medicine, Università del Piemonte Orientale UPO, Novara, Italy
- Division of Internal Medicine, “AOU Maggiore della Carità, Novara, Italy
| |
Collapse
|
13
|
Coculture With Ischemia/Reperfusion-Preconditioned Hepatocytes Improves Islet Function and Survival. Transplant Proc 2018; 50:3887-3894. [PMID: 30577282 DOI: 10.1016/j.transproceed.2018.07.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 07/04/2018] [Indexed: 12/11/2022]
Abstract
In clinical islet transplantation, hepatic ischemia and insufficient neovascularization of transplanted islets are barriers to islet survival and function. However, hepatocytes have a potency to protect themselves against ischemia. We hypothesized that ischemia/reperfusion preconditioning (IRP) of hepatocytes might beneficially affect islet cells in a coculture system. Primary islets were cocultured with primary hepatocytes, and hepatocyte IRP was conducted by subjecting cells to hypoxic conditions for single 15-minute/30-minute hypoxia, or 2 tandem 15-minute/30-minute hypoxic treatments (hypoxic-normoxic-hypoxic). We show that gene expression levels of insulin-like growth factor 1 (IGF-1), hepatocyte growth factor (HGF), transforming growth factor-α (TGF-α), and TGF-β1 in hepatocytes were increased by IRP. IRP hepatocytes secreted hepatocyte growth factor and insulin-like growth factor-1. Coculture of islets with IRP hepatocytes enhanced islet insulin secretion in glucose challenge test and expression of the survival-related gene Bcl-2 and the regenerating gene-1α (Reg-1α). Islets cocultured with the 30-minute double-IRP hepatocytes displayed significantly higher viability in the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and terminal deoxynucleotidyl transferase dUTP nick end labeling stain compared with that of islets subjected to 30 minutes of hypoxia. These results suggest that islet coculture with IRP hepatocytes can improve islet survival and insulin secretion.
Collapse
|
14
|
Cao Z, Ye T, Sun Y, Ji G, Shido K, Chen Y, Luo L, Na F, Li X, Huang Z, Ko JL, Mittal V, Qiao L, Chen C, Martinez FJ, Rafii S, Ding BS. Targeting the vascular and perivascular niches as a regenerative therapy for lung and liver fibrosis. Sci Transl Med 2018; 9:9/405/eaai8710. [PMID: 28855398 DOI: 10.1126/scitranslmed.aai8710] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 01/30/2017] [Accepted: 07/18/2017] [Indexed: 02/05/2023]
Abstract
The regenerative capacity of lung and liver is sometimes impaired by chronic or overwhelming injury. Orthotopic transplantation of parenchymal stem cells to damaged organs might reinstate their self-repair ability. However, parenchymal cell engraftment is frequently hampered by the microenvironment in diseased recipient organs. We show that targeting both the vascular niche and perivascular fibroblasts establishes "hospitable soil" to foster the incorporation of "seed," in this case, the engraftment of parenchymal cells in injured organs. Specifically, ectopic induction of endothelial cell (EC)-expressed paracrine/angiocrine hepatocyte growth factor (HGF) and inhibition of perivascular NOX4 [NADPH (reduced form of nicotinamide adenine dinucleotide phosphate) oxidase 4] synergistically enabled reconstitution of mouse and human parenchymal cells in damaged organs. Reciprocally, genetic knockout of Hgf in mouse ECs (HgfiΔEC/iΔEC) aberrantly up-regulated perivascular NOX4 during liver and lung regeneration. Dysregulated HGF and NOX4 pathways subverted the function of vascular and perivascular cells from an epithelially inductive niche to a microenvironment that inhibited parenchymal reconstitution. Perivascular NOX4 induction in HgfiΔEC/iΔEC mice recapitulated the phenotype of human and mouse liver and lung fibrosis. Consequently, EC-directed HGF and NOX4 inhibitor GKT137831 stimulated regenerative integration of mouse and human parenchymal cells in chronically injured lung and liver. Our data suggest that targeting dysfunctional perivascular and vascular cells in diseased organs can bypass fibrosis and enable reparative cell engraftment to reinstate lung and liver regeneration.
Collapse
Affiliation(s)
- Zhongwei Cao
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China. .,Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Tinghong Ye
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Yue Sun
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Gaili Ji
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Koji Shido
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Yutian Chen
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Lin Luo
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China.,West China Hospital, Sichuan University, China
| | - Feifei Na
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China.,West China Hospital, Sichuan University, China
| | - Xiaoyan Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Zhen Huang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Jane L Ko
- Department of Biological Sciences, Seton Hall University, South Orange, NJ 07079, USA
| | - Vivek Mittal
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, NY 10065, USA
| | - Lina Qiao
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Chong Chen
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China.,West China Hospital, Sichuan University, China
| | - Fernando J Martinez
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Shahin Rafii
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Bi-Sen Ding
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China. .,Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
15
|
Cordero-Espinoza L, Huch M. The balancing act of the liver: tissue regeneration versus fibrosis. J Clin Invest 2018; 128:85-96. [PMID: 29293095 DOI: 10.1172/jci93562] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Epithelial cell loss alters a tissue's optimal function and awakens evolutionarily adapted healing mechanisms to reestablish homeostasis. Although adult mammalian organs have a limited regeneration potential, the liver stands out as one remarkable exception. Following injury, the liver mounts a dynamic multicellular response wherein stromal cells are activated in situ and/or recruited from the bloodstream, the extracellular matrix (ECM) is remodeled, and epithelial cells expand to replenish their lost numbers. Chronic damage makes this response persistent instead of transient, tipping the system into an abnormal steady state known as fibrosis, in which ECM accumulates excessively and tissue function degenerates. Here we explore the cellular and molecular switches that balance hepatic regeneration and fibrosis, with a focus on uncovering avenues of disease modeling and therapeutic intervention.
Collapse
|
16
|
Yu J, Zhang W, Qian H, Tang H, Lin W, Lu B. SOCS1 regulates hepatic regenerative response and provides prognostic makers for acute obstructive cholangitis. Sci Rep 2017; 7:9482. [PMID: 28842621 PMCID: PMC5573403 DOI: 10.1038/s41598-017-09865-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 07/31/2017] [Indexed: 12/31/2022] Open
Abstract
Acute obstructive cholangitis (AOC) is a common and severe infectious diseases that occurs in an obstructed biliary system. The suppressors of cytokine signaling (SOCS) family include well-known negative regulators of cytokine receptor signaling. However, few studies have been conducted to determine their function in AOC. In this study, we showed that SOCS1 expression aberrantly changed and was associated with AOC prognosis in rat models. Decreased SOCS1 expression enhances regenerative response after biliary drainage (BD) resulting from AOC by upregulating hepatocyte growth factor (HGF) signaling. To detect SOCS1 expression in the liver less invasively and to predict the prognosis for AOC after BD, miR-221 and miR-222 were investigated. Ectopic SOCS1 expression indirectly decreases miR-221/222 expression through Met in vitro. An inverse correlation between SOCS1 expression and miR-221/222 expression in liver tissue or in serum was verified in rats. Serum from AOC patients showed that lower expression of circulating miR-221/222 after endoscopic nasobiliary drainage was associated with delayed restoration of liver function. Our results showed that SOCS1 regulates hepatic regenerative response, and indirectly detecting downstream molecules, such as miR-221/222, may provide prognostic makers for AOC.
Collapse
Affiliation(s)
- Jianhua Yu
- Department of Hepatobiliary Surgery, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing, China
| | - Weiguang Zhang
- Department of Molecular Medicine and Clinical Laboratory, Shaoxing Second Hospital, Shaoxing, China
| | - Hongwei Qian
- Department of Hepatobiliary Surgery, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing, China
| | - Haijun Tang
- Department of Hepatobiliary Surgery, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing, China
| | - Weiguo Lin
- Department of Hepatobiliary Surgery, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing, China
| | - Baochun Lu
- Department of Hepatobiliary Surgery, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing, China.
| |
Collapse
|
17
|
Vasanthan KS, Subramanian A, Krishnan UM, Sethuraman S. Development of Porous Hydrogel Scaffolds with Multiple Cues for Liver Tissue Engineering. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2017. [DOI: 10.1007/s40883-017-0034-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
18
|
Abstract
In vitro three-dimensional (3D) cultures are emerging as novel systems with which to study tissue development, organogenesis and stem cell behavior ex vivo. When grown in a 3D environment, embryonic stem cells (ESCs) self-organize into organoids and acquire the right tissue patterning to develop into several endoderm- and ectoderm-derived tissues, mimicking their in vivo counterparts. Tissue-resident adult stem cells (AdSCs) also form organoids when grown in 3D and can be propagated in vitro for long periods of time. In this Review, we discuss recent advances in the generation of pluripotent stem cell- and AdSC-derived organoids, highlighting their potential for enhancing our understanding of human development. We will also explore how this new culture system allows disease modeling and gene repair for a personalized regenerative medicine approach.
Collapse
Affiliation(s)
- Meritxell Huch
- Wellcome Trust/Cancer Research UK Gurdon Institute, Henry Wellcome Building of Cancer and Developmental Biology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge CB2 1QR, UK Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK
| | - Bon-Kyoung Koo
- Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge CB2 1QR, UK Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| |
Collapse
|
19
|
Regulation of soluble neuropilin 1, an endogenous angiogenesis inhibitor, in liver development and regeneration. Pathology 2015; 46:416-23. [PMID: 24977735 DOI: 10.1097/pat.0000000000000121] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Neuropilin-1 (NRP1) is a receptor for vascular endothelial growth factor (VEGF). A soluble isoform of Nrp1 (sNrp1) has not been described in the mouse. Our goal was to examine the expression of mouse sNrp1 during liver development and regeneration.sNrp1 was cloned from mouse liver. The expression of sNrp1 and VEGF was examined in mouse liver during post-natal development and regeneration using northern blot, western blot, in situ hybridisation, and immunohistochemical analyses. HGF/NRP1 binding was examined in vitro.A novel 588-amino acid sNrp1 isoform was found to contain the ligand binding regions of Nrp1. The adult liver expressed more sNrp1 than full-length Nrp1. In vivo, hepatocytes constitutively expressed VEGF and sNrp1 in the quiescent state. sNrp1 was highly up-regulated at P20, a time point coinciding with a plateau in liver and body weights. Following hepatectomy, endogenous levels of sNrp1 decreased during the rapid growth phase, and VEGF levels were highest just prior to and during the angiogenic phase. sNrp1 levels again rose 5-10 days post-hepatectomy, presumably to control regeneration. HGF protein bound NRP1 and binding was competed with sNRP1.We cloned a novel mouse sNrp1 isoform from liver and provide evidence that this endogenous angiogenesis inhibitor may regulate VEGF or HGF bioavailability during normal physiological growth and development as well as during liver regeneration.
Collapse
|
20
|
Palus M, Formanová P, Salát J, Žampachová E, Elsterová J, Růžek D. Analysis of serum levels of cytokines, chemokines, growth factors, and monoamine neurotransmitters in patients with tick-borne encephalitis: identification of novel inflammatory markers with implications for pathogenesis. J Med Virol 2015; 87:885-92. [PMID: 25675945 DOI: 10.1002/jmv.24140] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2014] [Indexed: 12/30/2022]
Abstract
Tick-borne encephalitis (TBE) is a leading human neuroinfection in Europe and northeastern Asia. However, the pathophysiology of TBE is not understood completely. This study sought to determine the specific serum mediators that are associated with acute TBE. The levels of 30 cytokines, chemokines, and growth factors were measured in serum samples from 87 patients with clinically and serologically confirmed acute TBE and from 32 control subjects using the Cytokine Human Magnetic 30-Plex Panel for the Luminex platform. Serum levels of the monoamine neurotransmitters serotonin, dopamine, and noradrenaline were measured via enzyme-linked immunosorbent assay. TBE virus infection elicited increased levels of the pro-inflammatory cytokines interleukin (IL)-6, IL-8, and IL-12. TBE patients had higher IL-12:IL-4 and IL-12:IL-10 ratios than control patients, reflecting the global pro-inflammatory cytokine balance. Serum levels of the monoamine neurotransmitters serotonin, dopamine, and noradrenaline were significantly lower in TBE patients than in the control group. Most interestingly, increased levels of hepatocyte growth factor and vascular endothelial growth factor were observed in TBE patients; these proteins may be novel and mechanistically important inflammatory biomarkers of TBE.
Collapse
Affiliation(s)
- Martin Palus
- Department of Virology, Veterinary Research Institute, Brno, Czech Republic; Laboratory of Arbovirology, Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, České Budějovice, Czech Republic; Department of Medical Biology, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | | | | | | | | | | |
Collapse
|
21
|
Abednazari H, Brudin L, Almroth G, Nilsson I, Nayeri F. Hepatocyte Growth Factor Is a Reliable Marker for Efficient Anti-Bacterial Therapy within the First Day of Treatment. ACTA ACUST UNITED AC 2014. [DOI: 10.4236/abb.2014.510096] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
22
|
Heynen GJJE, Fonfara A, Bernards R. Resistance to targeted cancer drugs through hepatocyte growth factor signaling. Cell Cycle 2014; 13:3808-17. [PMID: 25426675 PMCID: PMC4615055 DOI: 10.4161/15384101.2014.988033] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 11/11/2014] [Indexed: 01/22/2023] Open
Abstract
Cancer therapeutics that target a signaling pathway to which the cancer cells are addicted can deliver dramatic initial responses, but resistance is nearly always inevitable. A variety of mechanisms that cancer cells employ to escape from targeted cancer drugs have been described. We review here the role of Hepatocyte Growth Factor (HGF) and its receptor MET in drug resistance. We present data demonstrating that HGF can confer resistance to a number of kinase inhibitors in a variety of cancer cell lines and discuss our results in relation to the findings of others. Together, these data point at a major role for HGF/MET signaling in resistance to a variety of targeted cancer drugs.
Collapse
Affiliation(s)
- Guus JJE Heynen
- Division of Molecular Carcinogenesis and Cancer Genomics Netherlands; The Netherlands Cancer Institute; Amsterdam, The Netherlands
| | - Aldona Fonfara
- Division of Molecular Carcinogenesis and Cancer Genomics Netherlands; The Netherlands Cancer Institute; Amsterdam, The Netherlands
| | - René Bernards
- Division of Molecular Carcinogenesis and Cancer Genomics Netherlands; The Netherlands Cancer Institute; Amsterdam, The Netherlands
| |
Collapse
|
23
|
Wang H, Zhao T, Xu F, Li Y, Wu M, Zhu D, Cong X, Liu Y. How important is differentiation in the therapeutic effect of mesenchymal stromal cells in liver disease? Cytotherapy 2013; 16:309-18. [PMID: 24239106 DOI: 10.1016/j.jcyt.2013.07.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 07/23/2013] [Accepted: 07/29/2013] [Indexed: 12/17/2022]
Abstract
BACKGROUND AIMS The protocols for differentiation of hepatocyte-like cells (HLCs) from mesenchymal stromal cells (MSCs) have been well established. Previous data have shown that MSCs and their derived HLCs were able to engraft injured liver and alleviate injuries induced by carbon tetrachloride. The goal of the current study was to determine the differences of MSCs and their derived HLCs in terms of therapeutic functions in liver diseases. METHODS After hepatic differentiation of umbilical cord-derived MSCs in vitro, we detected both MSC and HLC expressions of adhesion molecules and chemokine receptor CXCR4 by flow cytometry; immunosuppressive potential and hepatocyte growth factor expression were determined by means of enzyme-linked immunosorbent assay. We compared the therapeutic effect for fulminant hepatic failure in a mouse model. RESULTS MSC-derived-HLCs expressed lower levels of hepatocyte growth factor, accompanied by impaired immunosuppression in comparison with MSCs. Furthermore, undifferentiated MSCs showed rescuing potentials superior to those in HLCs for the treatment of fulminant hepatic failure. CONCLUSIONS After differentiation, HLCs lost several major properties in comparison with undifferentiated MSCs, which are beneficial for their application in liver diseases. Undifferentiated MSCs may be more appropriate than are HLCs for the treatment of liver diseases.
Collapse
Affiliation(s)
- Hanyu Wang
- Alliancells Institute of Stem Cells and Translational Regenerative Medicine, Tianjin, China
| | - Tingting Zhao
- Alliancells Institute of Stem Cells and Translational Regenerative Medicine, Tianjin, China
| | - Fang Xu
- Alliancells Institute of Stem Cells and Translational Regenerative Medicine, Tianjin, China
| | - Yan Li
- Alliancells Institute of Stem Cells and Translational Regenerative Medicine, Tianjin, China
| | - Mingyuan Wu
- Alliancells Institute of Stem Cells and Translational Regenerative Medicine, Tianjin, China; Beijing Alliancells-PuRui Bioscience Co, Ltd, Beijing, China; University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Delin Zhu
- Alliancells Institute of Stem Cells and Translational Regenerative Medicine, Tianjin, China
| | - Xiuli Cong
- Alliancells Institute of Stem Cells and Translational Regenerative Medicine, Tianjin, China; University of Florida, Department of Medicine, Gainesville, Florida, USA
| | - Yongjun Liu
- Alliancells Institute of Stem Cells and Translational Regenerative Medicine, Tianjin, China.
| |
Collapse
|
24
|
Mungunsukh O, Day RM. Transforming growth factor-β1 selectively inhibits hepatocyte growth factor expression via a micro-RNA-199-dependent posttranscriptional mechanism. Mol Biol Cell 2013; 24:2088-97. [PMID: 23657814 PMCID: PMC3694793 DOI: 10.1091/mbc.e13-01-0017] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Hepatocyte growth factor (HGF) is a multipotent endogenous repair factor. The profibrotic cytokine transforming growth factor (TGF)-β1 inhibits HGF expression by a micro-RNA-199 (miR-199)-dependent posttranscriptional mechanism. In contrast, NK2, a truncated isoform of HGF that inhibits normal repair, is protected from TGF-β1–induced downregulation by miR-199. Hepatocyte growth factor (HGF) is a multipotent endogenous repair factor secreted primarily by mesenchymal cells with effects on cells expressing its receptor, Met. HGF promotes normal tissue regeneration and inhibits fibrotic remodeling in part by promoting proliferation and migration of endothelial and epithelial cells and protecting these cells from apoptosis. HGF also inhibits myofibroblast proliferation. The profibrotic cytokine transforming growth factor beta 1 (TGF-β1) suppresses HGF expression but not the expression of NK2, an HGF splice variant that antagonizes HGF-induced proliferation. We investigated the mechanism for differential regulation of HGF and NK2 by TGF-β1. TGF-β1 down-regulated HGF in primary human adult pulmonary fibroblasts (HLFb) and increased the expression of miR-199a-3p, a microRNA (miRNA) associated with fibrotic remodeling. HGF and NK2 contain completely different 3′ untranslated regions (UTRs), and we determined that miR-199a-3p targeted HGF mRNA for suppression but not NK2. A pre–miR-199 mimic inhibited the expression of a luciferase reporter harboring the HGF 3′ UTR but not a pmirGLO reporter containing the NK2 3′ UTR. In contrast, an anti-miRNA inhibitor specific for miR-199a-3p prevented TGF-β1–induced reduction of both HGF mRNA and HGF protein secretion. Taken together, these findings demonstrate that HGF is distinctly regulated at the posttranscriptional level from its antagonist NK2.
Collapse
Affiliation(s)
- Ognoon Mungunsukh
- Department of Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | | |
Collapse
|
25
|
The Effect of Elephantopus scaber L. on Liver Regeneration after Partial Hepatectomy. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:369180. [PMID: 23365606 PMCID: PMC3556435 DOI: 10.1155/2013/369180] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Revised: 12/04/2012] [Accepted: 12/04/2012] [Indexed: 11/18/2022]
Abstract
Liver regeneration after partial hepatectomy (PHx) is a physiological response for maintaining homeostasis. The aim of this study is to investigate effects of Elephantopus scaber L.- (ESL-) induced liver regeneration on growth factors (HGF and IGF-1), cell cycle regulation, and apoptosis suppressed. In this study, we fed five Chinese medicinal herbs (1 g/kg/day), Codonopsis pilosula (CP, Dangshen), Salvia miltiorrhiza Bunge (SMB, Danshen,), Bupleurum kasi (BK, Chaihu), Elephantopus scaber L. (ESL, Teng-Khia-U), and Silymarin (Sm, 25 mg/kg) for 7 days to male Spraue-Dawley rats. Then surgical 2/3 PHx was conducted and liver regeneration mechanisms were estimated on the following 24 hrs and 72 hrs. The activities of growth factors (HGF and IGF-I) and cell cycle proteins were measured by Western blot and RT-PCR. Histological analysis and apoptosis were detected by H&E stain and TUNEL. The results showed that extraction of Elephantopus scaber L. (ESL) and Silymarin (Sm, positive control) were increased protein expression levels of HGF and IGF-1 which leads into cell cycle. These results suggest that the ESL plays a crucial role in cell cycle-induced liver regeneration and apoptosis. These results suggested that the ESL plays a crucial role in cell cycle-induced liver regeneration and suppressed hepatocytes apoptosis.
Collapse
|
26
|
MET/HGF Signaling Pathway in Ovarian Carcinoma: Clinical Implications and Future Direction. PATHOLOGY RESEARCH INTERNATIONAL 2012; 2012:960327. [PMID: 23320251 PMCID: PMC3540829 DOI: 10.1155/2012/960327] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/09/2012] [Accepted: 12/07/2012] [Indexed: 11/18/2022]
Abstract
The HGF/MET signaling pathway is abnormal in numerous cancers including ovarian cancer. MET is expressed in 70% of human cancer and it is overexpressed in 30% of ovarian cases and cancer cell lines. The HGF/MET pathway plays a role in the initiation and progression of ovarian cancer through the most distinctive biologic program known as “invasive growth” which is accomplished through a coordinated activation of cell motility, invasiveness, degradation of extracellular matrix, survival, and proliferation. Because of its ubiquitous role in cancer, the MET axis seems to be an attractive target for cancer therapy. Numerous HGF/MET pathway inhibitor compounds are already in use in clinical trials in various solid tumors. In this paper, we will discuss the HGF/MET pathway in ovarian cancer, its clinical significance, and its potential use as a target therapy in the future.
Collapse
|
27
|
Overexpression of HGF transgene attenuates renal inflammatory mediators, Na(+)-ATPase activity and hypertension in spontaneously hypertensive rats. Biochim Biophys Acta Mol Basis Dis 2012; 1822:1590-9. [PMID: 22713485 DOI: 10.1016/j.bbadis.2012.06.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Revised: 06/10/2012] [Accepted: 06/11/2012] [Indexed: 12/24/2022]
Abstract
Renal inflammation and oxidative stress are constantly present in experimental hypertension. Since the spontaneously hypertensive rat (SHR) has reduced levels of hepatocyte growth factor (HGF), which suppresses the activation of the proinflammatory nuclear transcription factor kappa B (NF-κB), we speculated that HGF deficiency could play a key role in the pathogenesis of hypertension in the SHR. To test this hypothesis we increased HGF in the SHR by HGF gene delivery. We found that kidneys of 15-week-old SHR had an important reduction in HGF mRNA and protein expression. Adult SHRs were randomly assigned to receive weekly hydrodynamic injection (1mg/kg) of a naked plasmid containing human HGF (hHGF) gene associated with a cytomegalovirus promoter (pCMV-HGF) or empty vector (pcDNA3.1) during 6weeks. WKY rats treated with pcDNA3.1 and pCMV-HGF served as controls. The kidneys in the hypertensive SHR untreated and treated with the empty vector had increased NF-κB activation, elevated mRNA and protein expression of RANTES, MCP-1 and IL-6 and increased oxidative stress. Activity of Na(+)-ATPase was increased while activity of Na(+), K(+)-ATPase was normal. hHGF gene therapy normalized renal NF-κB activity, proinflammatory cytokines, antioxidant status (GSH, SOD and CAT), Na(+)-ATPase activity, reduced renal injury and ameliorated hypertension. Our results suggest that reduction in HGF production plays a major role in the pathogenesis of hypertension in the SHR and increasing HGF is a potential therapeutic target in the treatment of hypertension.
Collapse
|
28
|
Joo D, Kim J, Lee J, Kim Y, Fang Y, Jeong J, Kim M, Huh K. Impact of Coculture with Ischemic Preconditioned Hepatocellular Carcinoma Cell Line (Hep-G2) Cells on Insulin Secreting Function of Rat Insulin-secreting Cell Line (RIN-5F) Cells. Transplant Proc 2012; 44:1099-103. [DOI: 10.1016/j.transproceed.2012.02.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
29
|
Abshagen K, Eipel C, Vollmar B. A critical appraisal of the hemodynamic signal driving liver regeneration. Langenbecks Arch Surg 2012; 397:579-90. [PMID: 22311102 DOI: 10.1007/s00423-012-0913-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Accepted: 01/20/2012] [Indexed: 12/25/2022]
Abstract
BACKGROUND Many aspects of the signaling mechanisms involved in the initiation of hepatic regeneration are under current investigation. Nevertheless, the actual mechanisms switching liver regeneration on and off are still unknown. Hemodynamic changes in the liver following partial hepatectomy have been suggested to be a primary stimulus in triggering liver regeneration. Most of the new knowledge about the impact of hemodynamic changes on liver regeneration is both conceptually important and directly relevant to clinical problems. PURPOSE The purpose of this review is therefore to exclusively address the hemodynamic signal driving the liver regeneration process.
Collapse
Affiliation(s)
- Kerstin Abshagen
- Institute for Experimental Surgery, University of Rostock, 18055 Rostock, Germany.
| | | | | |
Collapse
|
30
|
Nakamura T, Sakai K, Nakamura T, Matsumoto K. Hepatocyte growth factor twenty years on: Much more than a growth factor. J Gastroenterol Hepatol 2011; 26 Suppl 1:188-202. [PMID: 21199531 DOI: 10.1111/j.1440-1746.2010.06549.x] [Citation(s) in RCA: 347] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Liver regeneration depends on the proliferation of mature hepatocytes. In the 1980s, the method for the cultivation of mature hepatocytes provided an opportunity for the discovery of hepatocyte growth factor (HGF) as a protein that is structurally and functionally different from other growth factors. In 1991, the scatter factor, tumor cytotoxic factor, and 3-D epithelial morphogen were identified as HGF, and Met tyrosine kinase was identified as the receptor for HGF. Thus, the connection of apparently unrelated research projects rapidly enriched the research on HGF in different fields. The HGF-Met pathway plays important roles in the embryonic development of the liver and the placenta, in the migration of myogenic precursor cells, and in epithelial morphogenesis. The use of tissue-specific knockout mice demonstrated that in mature tissues the HGF-Met pathway plays a critical role in tissue protection and regeneration, and in providing less susceptibility to chronic inflammation and fibrosis. In various injury and disease models, HGF promotes cell survival, regeneration of tissues, and suppresses and improves chronic inflammation and fibrosis. Drug development using HGF has been challenging, but extensive preclinical studies to address its therapeutic effects have provided significant results sufficient for the development of HGF as a biological drug in the regeneration-based therapy of diseases. Clinical trials using recombinant human HGF protein, or HGF genes, are in progress for the treatment of diseases.
Collapse
|
31
|
Factor VM, Seo D, Ishikawa T, Kaposi-Novak P, Marquardt JU, Andersen JB, Conner EA, Thorgeirsson SS. Loss of c-Met disrupts gene expression program required for G2/M progression during liver regeneration in mice. PLoS One 2010; 5. [PMID: 20862286 PMCID: PMC2940888 DOI: 10.1371/journal.pone.0012739] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2010] [Accepted: 08/15/2010] [Indexed: 12/13/2022] Open
Abstract
Background Previous work has established that HGF/c-Met signaling plays a pivotal role in regulating the onset of S phase following partial hepatectomy (PH). In this study, we used Metfl/fl;Alb-Cre+/− conditional knockout mice to determine the effects of c-Met dysfunction in hepatocytes on kinetics of liver regeneration. Methodology/Principal Finding The priming events appeared to be intact in Metfl/fl;Alb-Cre+/− livers. Up-regulation of stress response (MAFK, IKBZ, SOCS3) and early growth response (c-Myc, c-Jun, c-Fos, DUSP1 and 6) genes as assessed by RT-qPCR and/or microarray profiling was unchanged. This was consistent with an early induction of MAPK/Erk and STAT3. However, after a successful completion of the first round of DNA replication, c-Met deficient hepatocytes were blocked in early/mid G2 phase as shown by staining with phosphorylated form of histone H3. Furthermore, loss of c-Met in hepatocytes diminished the subsequent G1/S progression and delayed liver recovery after partial hepatectomy. Upstream signaling pathways involved in the blockage of G2/M transition included lack of persistent Erk1/2 activation and inability to up-regulate the levels of Cdk1, Plk1, Aurora A and B, and Mad2 along with a defective histone 3 phosphorylation and lack of chromatin condensation. Continuous supplementation with EGF in vitro increased proliferation of Metfl/fl;Alb-Cre+/− primary hepatocytes and partially restored expression levels of mitotic cell cycle regulators albeit to a lesser degree as compared to control cultures. Conclusion/Significance In conclusion, our results assign a novel non-redundant function for HGF/c-Met signaling in regulation of G2/M gene expression program via maintaining a persistent Erk1/2 activation throughout liver regeneration.
Collapse
Affiliation(s)
- Valentina M. Factor
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Daekwan Seo
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Tsuyoshi Ishikawa
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Pal Kaposi-Novak
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jens U. Marquardt
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jesper B. Andersen
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Elizabeth A. Conner
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Snorri S. Thorgeirsson
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
32
|
Salehi Z, Rajaei F. Expression of hepatocyte growth factor in the serum and cerebrospinal fluid of patients with Parkinson's disease. J Clin Neurosci 2010; 17:1553-6. [PMID: 20829049 DOI: 10.1016/j.jocn.2010.04.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2010] [Revised: 04/22/2010] [Accepted: 04/25/2010] [Indexed: 01/15/2023]
Abstract
Hepatocyte growth factor (HGF), also known as scatter factor, promotes the survival and migration of immature neurons. The HGF receptor c-Met is expressed in neurons. HGF plays an important role as a neurotrophic factor in the brain. HGF is produced by a wide variety of cells and is found in many physiological fluids, including serum and cerebrospinal fluid (CSF). Since CSF is in contact with the extracellular space of the brain, biochemical brain modifications are, to some extent, reflected in the CSF, and peptide and growth factors in the CSF can be used as biomarkers of disease. In this study, CSF and serum HGF concentrations were measured in patients with Parkinson's disease. The study population comprised 33 patients with Parkinson's disease and 38 normal controls. Western blot analysis using an anti-HGF antibody confirmed the presence of HGF in serum and CSF. No significant changes in serum HGF were observed in this study. However, CSF HGF expression was higher in patients with Parkinson's disease than in controls (p < 0.001). This finding indicates that HGF may be involved in the pathophysiology of Parkinson's disease.
Collapse
Affiliation(s)
- Zivar Salehi
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Iran.
| | | |
Collapse
|
33
|
Lindsay CD. Novel therapeutic strategies for acute lung injury induced by lung damaging agents: the potential role of growth factors as treatment options. Hum Exp Toxicol 2010; 30:701-24. [PMID: 20621953 DOI: 10.1177/0960327110376982] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The increasing threat from terrorism has brought attention to the possible use of toxic industrial compounds (TICs) and other lung-damaging agents as weapons against civilian populations. The way in which these agents could be used favours the development of generic countermeasures. Improved medical countermeasures would increase survivability and improve the quality of recovery of lung damaged casualties. It is evident that there is a dearth of therapeutic regimes available to treat those forms of lung damage that currently require intensive care management. It is quite possible that mass casualties from a terrorist incident or major industrial accident involving the release of large quantities of inhaled TICs would place a severe burden on already scarce intensive care facilities. The development of effective pharmacological approaches to assist the recovery of casualties suffering from acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) may improve the prognosis of such patients (which is currently poor) and would ideally be used as a means of preventing subjects from developing the pulmonary oedema characteristic of ALI/ARDS. Many promising candidate pharmacological treatments have been evaluated for the treatment of ALI/ARDS, but their clinical value is often debatable. Thus, despite improvements in ventilation strategies, pharmacological intervention for ALI/ARDS remains problematical. A new approach is clearly required for the treatment of patients with severely compromised lungs. Whilst the pathology of ALI/ARDS associated with exposure to a variety of agents is complex, numerous experimental studies suggest that generic therapeutic intervention directed at approaches that aim to upregulate repair of the damaged alveolar blood/air barrier of the lung may be of value, particularly with respect to chemical-induced injury. To this end, keratinocyte growth factor (KGF), epithelial growth factor (EGF) and basic fibroblast growth factor (bFGF) are emerging as the most important candidates. Hepatocyte growth factor (HGF) does not have epithelial specificity for lung tissue. However, the enhanced effects of combinations of growth factors, such as the synergistic effect of HGF upon vascular endothelial growth factor (VEGF)-mediated endothelial cell activity, and the combined effect of HGF and KGF in tissue repair should be investigated, particularly as the latter pair of growth factors are frequently implicated in processes associated with the repair of lung damage. Synergistic interactions also occur between trefoil factor family (TFF) peptides and growth factors such as EGF. TFF peptides are most likely to be of value as a short term therapeutic intervention strategy in stimulating epithelial spreading activities which allow damaged mucosal surfaces to be rapidly covered by epithelial cells.
Collapse
Affiliation(s)
- Christopher D Lindsay
- Biomedical Sciences Department, Defence Science and Technology Laboratory, Porton Down, Salisbury, Wiltshire, UK.
| |
Collapse
|
34
|
Chen G, Gingerich J, Soper L, Douglas GR, White PA. Induction of lacZ mutations in MutaMouse primary hepatocytes. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2010; 51:330-337. [PMID: 19953605 PMCID: PMC2959491 DOI: 10.1002/em.20540] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2009] [Revised: 09/18/2009] [Accepted: 09/17/2009] [Indexed: 05/28/2023]
Abstract
We have developed an in vitro mutation assay using primary hepatocytes from the transgenic MutaMouse. Primary hepatocytes were isolated using a two-step perfusion method with purification by Percoll, cultured, and treated with benzo[a]pyrene (BaP), 2-amino-1-methyl-6-phenyl- imidazo[4,5-b]pyridine (PhIP), 3-nitrobenzoanthrone (3-NBA), and cigarette smoke condensate (CSC). The mean lacZ mutant frequency (MF) for the solvent control was approximately twofold greater than the spontaneous MF observed in liver tissue. A concentration-dependent increase in MF (up to 3.7-fold above control) was observed following exposure to BaP. Fourfold and twofold increases in mutant frequency were observed for 3-NBA and PhIP exposures, respectively, without the addition of any exogenous metabolic activation. A slight but statistically significant increase in lacZ MF was observed for CSC, but only at the lowest concentration. This is the first report demonstrating that mutations can be detected in cultured primary hepatocytes from MutaMouse. The preliminary results presented suggest that the MutaMouse primary hepatocyte mutagenicity assay can be used as a cost-effective tool for screening of environmental mutagens and therapeutic products.
Collapse
Affiliation(s)
- Guosheng Chen
- Environmental Health Sciences and Research Bureau, Research and Radiation Directorate, Health Canada, Ottawa, Ontario, Canada
| | | | | | | | | |
Collapse
|
35
|
Zhong N, Huang F, Tan G, Jiao J, Lin Y, Wang C, Wang H, Zhou S, Huang Y, Chen F, Lin Y. Effect of hepatocyte growth factor signaling pathway activation on Plasmodium berghei infection. ASIAN PAC J TROP MED 2010. [DOI: 10.1016/s1995-7645(10)60001-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
36
|
Ijima H, Kubo T, Hou YT. Primary rat hepatocytes form spheroids on hepatocyte growth factor/heparin-immobilized collagen film and maintain high albumin production. Biochem Eng J 2009. [DOI: 10.1016/j.bej.2009.05.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
37
|
Schievenbusch S, Strack I, Scheffler M, Wennhold K, Maurer J, Nischt R, Dienes HP, Odenthal M. Profiling of anti-fibrotic signaling by hepatocyte growth factor in renal fibroblasts. Biochem Biophys Res Commun 2009; 385:55-61. [PMID: 19426716 DOI: 10.1016/j.bbrc.2009.05.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2009] [Accepted: 05/03/2009] [Indexed: 01/16/2023]
Abstract
Hepatocyte growth factor (HGF) is a multifunctional growth factor affecting cell proliferation and differentiation. Due to its mitogenic potential, HGF plays an important role in tubular repair and regeneration after acute renal injury. However, recent reports have shown that HGF also acts as an anti-inflammatory and anti-fibrotic factor, affecting various cell types such as renal fibroblasts and triggering tubulointerstitial fibrosis of the kidney. The present study provides evidence that HGF stimulation of renal fibroblasts results in the activation of both the Erk1/2 and the Akt pathways. As previously shown, Erk1/2 phosphorylation results in Smad-linker phosphorylation, thereby antagonizing cellular signals induced by TGFbeta. By siRNA mediated silencing of the Erk1/2-Smad linkage, however, we now demonstrate that Akt signaling acts as an auxiliary pathway responsible for the anti-fibrotic effects of HGF. In order to define the anti-fibrotic function of HGF we performed comprehensive expression profiling of HGF-stimulated renal fibroblasts by microarray hybridization. Functional cluster analyses and quantitative PCR assays indicate that the HGF-stimulated pathways transfer the anti-fibrotic effects in renal interstitial fibroblasts by reducing expression of extracellular matrix proteins, various chemokines, and members of the CCN family.
Collapse
Affiliation(s)
- Stephanie Schievenbusch
- Institute for Pathology, University Hospital Cologne, Kerpener Str. 62, 50924 Koeln, Germany
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Ishii K, Yoshida Y, Akechi Y, Sakabe T, Nishio R, Ikeda R, Terabayashi K, Matsumi Y, Gonda K, Okamoto H, Takubo K, Tajima F, Tsuchiya H, Hoshikawa Y, Kurimasa A, Umezawa A, Shiota G. Hepatic differentiation of human bone marrow-derived mesenchymal stem cells by tetracycline-regulated hepatocyte nuclear factor 3beta. Hepatology 2008; 48:597-606. [PMID: 18666263 DOI: 10.1002/hep.22362] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
UNLABELLED Human bone marrow-derived mesenchymal stem cells (BM-MSCs) are expected to be a potential source of cells for transplantation. Although recent reports have shown that isolated MSCs can differentiate into hepatocytes, the efficiency of differentiation is insufficient for therapeutic application. To circumvent this problem, it is necessary to understand the mechanisms of hepatic differentiation of human BM-MSCs. Hepatocyte nuclear factor 3beta (HNF3beta), a forkhead/winged helix transcription factor, is essential for liver development. In the present study, we established a tetracycline (Tet)-regulated expression system for HNF3beta in UE7T-13 BM-MSCs. HNF3beta expression significantly enhanced expression of albumin, alpha-fetoprotein (AFP), tyrosine amino transferase (TAT) and epithelial cell adhesion molecule (EpCAM) genes. The differentiated cells showed hepatocyte-specific functions including glycogen production and urea secretion. During treatment with the Tet-on system for 8 days, over 80% of UE7T-13 cells turned out to express albumin. Furthermore, the combination of Tet with basic fibroblast growth factor (bFGF) efficiently induced the genes such as albumin and TAT, which are associated with maturity of hepatocytes; however, it suppressed genes such as AFP and EpCAM, which are associated with immaturity of hepatocytes, suggesting that Tet-induced HNF3beta expression sensitizes BM-MSCs to bFGF signals. Finally, the results of the present study suggest that down-regulation of Wnt/beta-catenin signals caused by translocation of beta-catenin to cytoplasmic membrane is associated with hepatic differentiation of human BM-MSCs. CONCLUSION HNF3beta expression induced efficient differentiation of UE7T-13 human BM-MSCs.
Collapse
Affiliation(s)
- Kyoko Ishii
- Department of Genetic Medicine and Regenerative Therapeutics, Division of Molecular and Genetic Medicine, Graduate School of Medicine, Tottori University, Yonago, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Nayeri F, Nayeri T, Aili D, Brudin L, Liedberg B. Clinical impact of real-time evaluation of the biological activity and degradation of hepatocyte growth factor. Growth Factors 2008; 26:163-71. [PMID: 18569024 PMCID: PMC2562019 DOI: 10.1080/08977190802128083] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Hepatocyte growth factor (HGF) is essential for injury repair. Despite high HGF levels in chronic ulcers, up-regulation of HGF receptor in ulcer tissue and decreased biological activity of HGF in ulcer secretions have been observed. With a surface plasmon resonance-based method, we assessed the binding of HGF to antibodies, receptors, and the basement membrane and identified binding interactions that are indispensable for the biological activity of HGF. Recombinant HGF (rHGF) lots were tested for activity, structural integrity, and degradation, and the results were verified in an in vitro model of cell injury. Biologically active rHGF, as well as plasma from healthy volunteers, bound to heparan sulphate proteoglycan (HSPG) and to anti-HGF antibodies. Decreased binding to HSPG was the first event in rHGF degradation. This study established the feasibility of identifying patients with chronic inflammation who need exogenous HGF and of using ligand-binding assessment to evaluate rHGF lots for biological activity.
Collapse
Affiliation(s)
- Fariba Nayeri
- Division of Infectious Diseases, Faculty of Health Science, University Hospital, Linkoping, Sweden.
| | | | | | | | | |
Collapse
|
40
|
Frevert U, Späth GF, Yee H. Exoerythrocytic development of Plasmodium gallinaceum in the White Leghorn chicken. Int J Parasitol 2008; 38:655-72. [PMID: 18005972 PMCID: PMC2430052 DOI: 10.1016/j.ijpara.2007.09.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2007] [Revised: 09/22/2007] [Accepted: 09/27/2007] [Indexed: 01/22/2023]
Abstract
Plasmodium gallinaceum typically causes sub-clinical disease with low mortality in its primary host, the Indian jungle fowl Gallus sonnerati. Domestic chickens of European origin, however, are highly susceptible to this avian malaria parasite. Here we describe the development of P. gallinaceum in young White Leghorn chicks with emphasis on the primary exoerythrocytic phase of the infection. Using various regimens for infection, we found that P. gallinaceum induced a transient primary exoerythrocytic infection followed by a fulminant lethal erythrocytic phase. Prerequisite for the appearance of secondary exoerythrocytic stages was the development of a certain level of parasitaemia. Once established, secondary exoerythrocytic stages could be propagated from bird to bird for several generations without causing fatalities. Infected brains contained large secondary exoerythrocytic stages in capillary endothelia, while in the liver primary and secondary erythrocytic stages developed primarily in Kupffer cells and remained smaller. At later stages, livers exhibited focal hepatocyte necrosis, Kupffer cell hyperplasia, stellate cell proliferation, inflammatory cell infiltration and granuloma formation. Because P. gallinaceum selectively infected Kupffer cells in the liver and caused a histopathology strikingly similar to mammalian species, this avian Plasmodium species represents an evolutionarily closely related model for studies on the hepatic phase of mammalian malaria.
Collapse
Affiliation(s)
- Ute Frevert
- Department of Medical Parasitology, New York University School of Medicine, New York, NY 10010, USA.
| | | | | |
Collapse
|
41
|
Enoch S, Moseley R, Stephens P, Thomas D. The oral mucosa: a model of wound healing with reduced scarring. ACTA ACUST UNITED AC 2008. [DOI: 10.1111/j.1752-248x.2007.00005.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
42
|
Gong R, Rifai A, Ge Y, Chen S, Dworkin LD. Hepatocyte growth factor suppresses proinflammatory NFkappaB activation through GSK3beta inactivation in renal tubular epithelial cells. J Biol Chem 2008; 283:7401-10. [PMID: 18201972 DOI: 10.1074/jbc.m710396200] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Activation of NFkappaB is a fundamental cellular event central to all inflammatory diseases. Hepatocyte growth factor (HGF) ameliorates both acute and chronic inflammation in a multitude of organ systems through modulating NFkappaB activity; nevertheless, the exact molecular mechanism remains uncertain. Here we report that HGF through inactivation of GSK3beta suppresses NFkappaB p65 phosphorylation specifically at position Ser-468. The Ser-468 of RelA/p65 situates in a GSK3beta consensus motif and could be directly phosphorylated by GSK3beta both in vivo and in vitro, signifying Ser-468 of RelA/p65 as a putative substrate for GSK3beta. In addition, the C terminus of RelA/p65 harbors a highly conserved domain homologue of the consensus docking sequence for GSK3beta. Moreover, this domain was required for efficient phosphorylation of Ser-468 and was indispensable for the physical interaction between RelA/p65 and GSK3beta. HGF substantially intercepted this interaction by inactivating GSK3beta. Functionally, phosphorylation of Ser-468 of RelA/p65 was required for the induced expression of a particular subset of proinflammatory NFkappaB-dependent genes. Diminished phosphorylation at Ser-468 by HGF resulted in a gene-specific inhibition of these genes' expression. The action of HGF on proinflammatory NFkappaB activation was consistently mimicked by a selective GSK3beta inhibitor or GSK3beta knockdown by RNA interference but largely abrogated in cells expressing the mutant uninhibitable GSK3beta. Collectively, our findings suggest that HGF has a potent suppressive effect on NFkappaB activation, which is mediated by GSK3beta, an important signaling transducer controlling RelA/p65 phosphorylation specificity and directing the transcription of selective proinflammatory cytokines implicated in inflammatory kidney disease.
Collapse
Affiliation(s)
- Rujun Gong
- Division of Kidney Disease and Hypertension, Department of Medicine, Brown University School of Medicine, Provindence, Rhode Island 02903, USA.
| | | | | | | | | |
Collapse
|
43
|
Tsai HC, Chen YS, Lee SSJ, Lin YH, Wann SR, Sy CL, Liu YC. Hepatocyte growth factor level in cerebrospinal fluid as an additional marker in patient with drug-resistant streptococcus pneumoniae meningitis treated with linezolid. Clin Med Case Rep 2008; 1:53-5. [PMID: 24179347 PMCID: PMC3785359 DOI: 10.4137/ccrep.s759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Hepatocyte growth factor (HGF) is a multifunctional cytokine that has well-defined mitogenic, motogenic, and morphogenic functions on the epithelial cells. Strong increases of HGF concentrations in cerebrospinal fluid (CSF) are only present in patients with acute bacterial meningitis. We report a 15-year-old young man with drug resistant Streptococcus pneumoniae meningitis complicated with right 6th cranial nerve palsy. He presented with fever, headache and neck stiffness for 3 days and was treated with intravenous antimicrobial agents including linezolid and dexamethasone successfully. An association between CSF HGF concentrations and CSF proteins is observed (r = 0.897, p = 0.039. Pearson correlation test). This result showed that HGF level in CSF might act as an additional marker in patients with bacterial meningitis.
Collapse
Affiliation(s)
- Hung-Chin Tsai
- Section of Infectious Diseases, Department of Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan, National Yang-Ming University, Taipei, Taiwan, Republic of China. ; Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China
| | | | | | | | | | | | | |
Collapse
|
44
|
Yuan RH, Chen HL, Chen HL, Hsu MK, Lee PH, Chang MH. Attenuation of Kupffer Cell Function in Acute on Chronic Liver Injury Enhanced Engraftment of Transplanted Hepatocytes. World J Surg 2007; 31:1270-7; discussion 1278-9. [PMID: 17437157 DOI: 10.1007/s00268-007-9044-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND The present study was designed to elucidate the relationship of engraftment efficiency of transplanted cells and Kupffer cell function in mice with acute on chronic liver injury and acute liver injury. METHODS The recipient dipeptidyl peptidase IV knockout (DPPIV(-/-)) mice were divided into two groups: (1) the acute on chronic liver injury group (CCl(4)/APAP group) that received CCl(4) (1 ml/kg) twice a week for 4 weeks following one dose of acetaminophen (APAP), 600 mg/kg; (2) the acute liver injury group (APAP-only group) that received a single dose of APAP at 600 mg/kg. DPPIV(+/+) hepatocytes were transplanted 24 h after APAP intoxication. Engraftment efficiency was evaluated at day 7 and day 14 after transplantation. The tumor necrosis factor-alpha (TNF-alpha) mRNA expression level of Kupffer cells immediately before cell transplantation was compared between the two groups before and after lipopolysaccharide (LPS, 100 ng/ml) stimulation. RESULTS The number of transplanted cells and clusters in each 100x microscopic field were higher in the CCl(4)/APAP group at both day 7 (21.5 +/- 6.3 versus 8.3 +/- 4.0, p < 0.001; 14.9 +/- 4.6 versus 6.6 +/- 3.4, p < 0.001, respectively) and day 14 (17.3 +/- 4.4 versus 10.2 +/- 3.3, p = 0.001; 12.6 +/- 3.2 versus 7.9 +/- 1.6, p = 0.004, respectively). After LPS stimulation, the expression level of TNF-alpha was lower (175.7 +/- 54.6 versus 465.6 +/- 64.2, p = 0.002), and the increment of TNF-alpha expression was also less significant in the CCl(4)/APAP group (1.5-fold versus 6.5-fold, p = 0.014). CONCLUSIONS Chronic liver injury desensitized Kupffer cells and reduced TNF-alpha expression, two results that correlated with the increased engraftment of transplanted cells.
Collapse
Affiliation(s)
- Ray-Hwang Yuan
- Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
45
|
Nayeri F, Holmgren-Pettersson K, Perskvist N, Forsberg P, Peterson C, Sundqvist T. An in vitro model for assessment of the biological activity of hepatocyte growth factor. Growth Factors 2007; 25:33-40. [PMID: 17454148 DOI: 10.1080/08977190600997200] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Hepatocyte growth factor (HGF) is a multifunctional growth factor with potent wound-healing properties that functions in the healing of chronic injuries. However, there may be a loss of HGF activity in certain chronic cases; this might be indicated by the presence of high amounts of HGF in body fluids and by the elevated expression of the HGF receptor in tissue biopsies. In such cases, a reliable means of assessing the activity of endogenous HGF would be valuable in allowing clinicians to decide if treatment with HGF would be useful. In this study, we developed an in vitro wound assay that used a mouse skin epithelial cell line to evaluate the biological activity of HGF. We showed that HGF accelerated the motility of the epithelial cells in a dose-dependent fashion with high sensitivity and specificity. This in vitro assay might be used to determine the activity of both endogenous and recombinant HGF.
Collapse
Affiliation(s)
- Fariba Nayeri
- Division of Infectious Diseases, Faculty of Health Science, Linköping University, Linköping, SE-581 85, Sweden.
| | | | | | | | | | | |
Collapse
|
46
|
Harun N, Nikfarjam M, Muralidharan V, Christophi C. Liver regeneration stimulates tumor metastases. J Surg Res 2007; 138:284-90. [PMID: 17254608 DOI: 10.1016/j.jss.2006.06.024] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2006] [Revised: 06/09/2006] [Accepted: 06/22/2006] [Indexed: 12/11/2022]
Abstract
BACKGROUND Partial hepatectomy for patients with colorectal liver metastases is associated with a tumor recurrence rate approaching 80% post-resection. Different factors and phases associated with regeneration of the liver are implicated in tumor recurrence. This study investigates the effects of the early and late phases of liver regeneration and the impact of the degree of liver resection on stimulating tumor growth and metastasis. MATERIALS AND METHODS Groups of mice underwent partial hepatectomy (37% or 70%) and were then challenged with colorectal liver carcinoma (CRC) tumors immediately after liver resection (early and late phase effect) or 6 days post liver resection (late phase effect). Tumor growth, degree of proliferation, tumor morphology, and the presence of extrahepatic metastases were investigated 21 days post-tumor induction. RESULTS The late phase of liver regeneration plays a significant role in tumor stimulation and metastasis. The degree of hepatectomy also appears to be an important factor. The degree of hepatic resection significantly influences tumor growth and the extent of extrahepatic metastases, particularly in the lungs. CONCLUSIONS Elucidation of the processes involved in the late phase of liver regeneration may assist in the development and timing of adjuvant agents to minimize tumor recurrence during this phase.
Collapse
Affiliation(s)
- Nadia Harun
- Department of Surgery, University of Melbourne, Austin Hospital, Heidelberg, Victoria, Australia.
| | | | | | | |
Collapse
|
47
|
Chau GY, Lui WY, Chi CW, Chau YP, Li AF, Kao HL, Wu CW. Significance of serum hepatocyte growth factor levels in patients with hepatocellular carcinoma undergoing hepatic resection. Eur J Surg Oncol 2007; 34:333-8. [PMID: 17218078 DOI: 10.1016/j.ejso.2006.12.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2006] [Accepted: 12/05/2006] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Hepatocyte growth factor (HGF) is a potent hepatocyte mitogen and may stimulate the proliferation and invasiveness of human hepatocellular carcinoma (HCC) cells through the c-met receptor. This study evaluates the significance of serum HGF levels in patients undergoing HCC resection. STUDY DESIGN The peripheral and portal sera and HCC and non-tumorous tissues of 40 HCC patients, with tumor TNM stage I (n=12), II (n=17), and III (n=11) diseases, who underwent hepatic resection were prospectively collected. Serum HGF levels were determined by enzyme-linked immunosorbent assay. The c-met protein expressions were examined by immunohistochemistry. Median follow-up time was 69 months. RESULTS The prehepatectomy portal HGF levels (median, 622pg/mL) were significantly higher than peripheral HGF levels (564pg/mL) (P=0.026). The posthepatectomy portal HGF levels (699pg/mL) were significantly higher than prehepatectomy portal HGF levels (P<0.001). C-met expression was detected in 87.5% HCC and in 85.0% non-tumorous liver tissues. By Cox multivariate analysis, posthepatectomy portal HGF level >699pg/mL (P<0.001), multiple tumors (P=0.042), and TNM stages II (P=0.019) and III (P=0.009) were independent factors related with survival. Patients with a posthepatectomy portal HCG level >699pg/mL and with a positive c-met expression in HCC tissue have the worst survival. CONCLUSIONS In HCC patients, high peripheral and portal HGF serum levels related with poor prognosis after hepatic resection. Hepatocyte growth factor and c-met receptor can be targets of future HCC postoperative treatment.
Collapse
Affiliation(s)
- G-Y Chau
- Department of Surgery, Division of General Surgery, Taipei Veterans General Hospital, School of Medicine, National Yang-Ming University, No. 201, Shih-pai Road, Section 2, Shih-pai, Taipei 112, Taiwan.
| | | | | | | | | | | | | |
Collapse
|
48
|
Alvaro D, Mancino MG, Glaser S, Gaudio E, Marzioni M, Francis H, Alpini G. Proliferating cholangiocytes: a neuroendocrine compartment in the diseased liver. Gastroenterology 2007; 132:415-31. [PMID: 17241889 DOI: 10.1053/j.gastro.2006.07.023] [Citation(s) in RCA: 220] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2006] [Accepted: 07/12/2006] [Indexed: 12/16/2022]
Abstract
In the last 15 years, the intrahepatic biliary tree has become the object of extensive studies, which highlighted the extraordinary biologic properties of cholangiocytes involved in bile formation, proliferation, injury repair, fibrosis, angiogenesis, and regulation of blood flow. Proliferation is a "typical" property of cholangiocytes and is key as a mechanism of repair responsible for maintaining the integrity of the biliary tree. Cholangiocyte proliferation occurs virtually in all pathologic conditions of liver injury where it is associated with inflammation, regeneration, and repair, thus conditioning the evolution of liver damage. Interestingly, proliferating cholangiocytes acquire the phenotype of neuroendocrine cells, and secrete different cytokines, growth factors, neuropeptides, and hormones, which represent potential mechanisms for cross talk with other liver cells. Many studies suggest the generation of a neuroendocrine compartment in the injured liver, mostly constituted by cells with cholangiocyte features, which functionally conditions the progression of liver disease. These insights on cholangiocyte pathophysiology will provide new potential strategies for the management of chronic liver diseases. The purpose of this review is to summarize the recent findings on the mechanisms regulating cholangiocyte proliferation and the significance of the neuroendocrine regulation of cholangiocyte biology.
Collapse
Affiliation(s)
- Domenico Alvaro
- Division of Gastroenterology, Department of Clinical Medicine, University La Sapienza, via R. Rossellini 51, 00137 Rome, Italy.
| | | | | | | | | | | | | |
Collapse
|
49
|
Gong R, Rifai A, Dworkin LD. Hepatocyte growth factor suppresses acute renal inflammation by inhibition of endothelial E-selectin. Kidney Int 2006; 69:1166-74. [PMID: 16501492 PMCID: PMC7126584 DOI: 10.1038/sj.ki.5000246] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Vascular endothelial activation, marked by de novo expression of E-selectin, is an early and essential event in the process of leukocyte extravasation and inflammation. Evidence suggests that hepatocyte growth factor (HGF) ameliorates inflammation in animal models of renal disease, implying that HGF might inhibit specific components of the inflammatory response. This study examined the effect of HGF on endothelial E-selectin expression in acute inflammation induced by tumor necrosis factor (TNF)-α. In vitro, HGF suppressed TNF-α-induced cell surface expression of E-selectin in human umbilical vein endothelial cells (HUVEC) and inhibited E-selectin mediated monocytic adhesion to endothelial monolayers. HGF activated phosphatidylinositol 3-kinase (PI3K)–Akt that in turn inhibited its downstream transducer glycogen synthase kinase (GSK)3. Blockade of the PI3K–Akt pathway with specific inhibitors abrogated HGF induced inhibitory phosphorylation of GSK3 and suppression of E-selectin. In addition, selective inhibition of GSK3 activity by lithium suppressed TNF-α-induced E-selectin expression and monocytic adhesion, reminiscent of the action of HGF. Moreover, ectopic expression of an uninhibitable mutant GSK3β, in which the regulatory serine-9 is replaced by alanine, abolished HGF's suppressive effect on endothelial E-selectin. In vivo, administration of exogenous HGF reduced endothelial expression of E-selectin induced by bolus injection of TNF-α. This was associated with less sequestration of circulating fluorescence-labeled macrophages in the kidney. These findings suggest that HGF ameliorates acute renal inflammation in part by downregulating E-selectin mediated macrophage adhesion to the inflamed endothelium.
Collapse
Affiliation(s)
- R Gong
- Department of Medicine, Division of Renal Diseases, Brown University School of Medicine, Providence, Rhode Island 02903, USA.
| | | | | |
Collapse
|
50
|
Abednazari H, Xu J, Millinger E, Brudin L, Forsberg P, Nayeri F. Hepatocyte growth factor is a better indicator of therapeutic response than C-reactive protein within the first day of treatment in pneumonia. Chemotherapy 2006; 52:260-3. [PMID: 16899975 DOI: 10.1159/000094868] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2004] [Accepted: 09/06/2005] [Indexed: 11/19/2022]
Abstract
Acute bacterial infectious diseases are mostly treated empirically at admission before the culture results are available. According to the risk for serious complications in the case of therapeutic failure, it is important to evaluate the therapy results and change to a more appropriate antibiotic regime as soon as possible. In the present study, 40 patients with X-ray-verified community-acquired pneumonia were examined and blood specimens were collected before and within 24 h of treatment. Body temperature, C-reactive protein (CRP) and hepatocyte growth factor (HGF) were investigated. Thirty-two patients received an appropriate initial antibiotic therapy regarding clinical outcome, but in 8 patients the treatment was changed because of therapy failure. Changes of HGF levels after 18-24 h of treatment could predict the therapeutic results accurately in 38 of 40 cases (sensitivity 100%, specificity 94%, positive likelihood ratio 16.0). HGF was significantly better to predict therapy outcome than CRP (p < 0.0001).
Collapse
|