1
|
Elmi M, Dass JH, Dass CR. The Various Roles of PEDF in Cancer. Cancers (Basel) 2024; 16:510. [PMID: 38339261 PMCID: PMC10854708 DOI: 10.3390/cancers16030510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/16/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Pigment epithelium-derived factor (PEDF) is a natural immunomodulator, anti-inflammatory, anti-angiogenic, anti-tumour growth and anti-metastasis factor, which can enhance tumour response to PEDF but can also conversely have pro-cancerous effects. Inflammation is a major cause of cancer, and it has been proven that PEDF has anti-inflammatory properties. PEDF's functional activity can be investigated through measuring metastatic and metabolic biomarkers that will be discussed in this review.
Collapse
Affiliation(s)
- Mitra Elmi
- Curtin Medical School, Curtin University, Bentley, WA 6102, Australia; (M.E.); (J.H.D.)
- Curtin Health Innovation Research Institute, Curtin Medical School, Curtin University, Bentley, WA 6102, Australia
| | - Joshua H. Dass
- Curtin Medical School, Curtin University, Bentley, WA 6102, Australia; (M.E.); (J.H.D.)
- Sir Charles Gairdner Hospital, Nedlands, WA 6009, Australia
| | - Crispin R. Dass
- Curtin Medical School, Curtin University, Bentley, WA 6102, Australia; (M.E.); (J.H.D.)
- Curtin Health Innovation Research Institute, Curtin Medical School, Curtin University, Bentley, WA 6102, Australia
| |
Collapse
|
2
|
Brook N, Dharmarajan A, Chan A, Dass CR. Potential therapeutic role for pigment epithelium-derived factor in post-menopausal breast cancer bone metastasis. J Pharm Pharmacol 2023:7146711. [PMID: 37116213 DOI: 10.1093/jpp/rgad039] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 04/12/2023] [Indexed: 04/30/2023]
Abstract
OBJECTIVES This review discusses key oestrogens associated with the circulating pre- and post-menopausal milieu and how they may impact intratumoral oestrogen levels and breast cancer (BC) metastasis. It also identifies critical steps in BC metastasis to bone from the viewpoint of pigment epithelium-derived factor (PEDF) function, and discusses the role of several associated pro-metastatic biomarkers in BC bone metastasis. KEY FINDINGS PEDF is regulated by oestrogen in a number of oestrogen-sensitive tissues. Changes in circulating oestrogen levels associated with menopause may enhance the growth of BC bone metastases, leading to the establishment of a pre-metastatic niche. The establishment of such a pre-metastatic niche is driven by several key mediators, with pro-osteoclastic and pro-metastatic function which are upregulated by BC cells. These mediators appear to be regulated by oestrogen, as well as differentially affected by menopausal status. PEDF interacts with several pro-metastatic, pro-osteoclastic biomarkers, including C-X-C motif chemokine receptor 4 (CXCR4) and nuclear factor kappa B (NFκB) in BC bone metastasis. CONCLUSION Mediators such as CXCR4 and MT1-MMP underpin the ability of PEDF to function as an antimetastatic in other cancers such as osteosarcoma, highlighting the possibility that this serpin could be used as a therapeutic against BC metastasis in future.
Collapse
Affiliation(s)
- Naomi Brook
- Curtin Medical School, Curtin University, Bentley 6102, Australia
- Curtin Health Innovation Research Institute, Bentley 6102, Australia
| | - Arun Dharmarajan
- Curtin Medical School, Curtin University, Bentley 6102, Australia
- Curtin Health Innovation Research Institute, Bentley 6102, Australia
- Department of Biomedical Sciences, Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai 600116, India
| | - Arlene Chan
- Curtin Medical School, Curtin University, Bentley 6102, Australia
- Breast Cancer Research Centre-Western Australia, Hollywood Private Hospital, Nedlands 6009, Australia
| | - Crispin R Dass
- Curtin Medical School, Curtin University, Bentley 6102, Australia
- Curtin Health Innovation Research Institute, Bentley 6102, Australia
| |
Collapse
|
3
|
Cadamuro M, Romanzi A, Guido M, Sarcognato S, Cillo U, Gringeri E, Zanus G, Strazzabosco M, Simioni P, Villa E, Fabris L. Translational Value of Tumor-Associated Lymphangiogenesis in Cholangiocarcinoma. J Pers Med 2022; 12:jpm12071086. [PMID: 35887583 PMCID: PMC9324584 DOI: 10.3390/jpm12071086] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/23/2022] [Accepted: 06/29/2022] [Indexed: 11/16/2022] Open
Abstract
The prognosis of cholangiocarcinoma remains poor in spite of the advances in immunotherapy and molecular profiling, which has led to the identification of several targetable genetic alterations. Surgical procedures, including both liver resection and liver transplantation, still represent the treatment with the best curative potential, though the outcomes are significantly compromised by the early development of lymph node metastases. Progression of lymphatic metastasis from the primary tumor to tumor-draining lymph nodes is mediated by tumor-associated lymphangiogenesis, a topic largely overlooked until recently. Recent findings highlight tumor-associated lymphangiogenesis as paradigmatic of the role played by the tumor microenvironment in sustaining cholangiocarcinoma invasiveness and progression. This study reviews the current knowledge about the intercellular signaling and molecular mechanism of tumor-associated lymphangiogenesis in cholangiocarcinoma in the hope of identifying novel therapeutic targets to halt a process that often limits the success of the few available treatments.
Collapse
Affiliation(s)
| | - Adriana Romanzi
- Gastroenterology Unit, Department of Medical Specialties, University of Modena & Reggio Emilia and Modena University-Hospital, 41124 Modena, Italy;
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Maria Guido
- Department of Pathology, Azienda ULSS2 Marca Trevigiana, 31100 Treviso, Italy; (M.G.); (S.S.)
- Department of Medicine (DIMED), University of Padua, 35122 Padua, Italy;
| | - Samantha Sarcognato
- Department of Pathology, Azienda ULSS2 Marca Trevigiana, 31100 Treviso, Italy; (M.G.); (S.S.)
| | - Umberto Cillo
- Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padua, 35122 Padua, Italy; (U.C.); (E.G.); (G.Z.)
| | - Enrico Gringeri
- Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padua, 35122 Padua, Italy; (U.C.); (E.G.); (G.Z.)
| | - Giacomo Zanus
- Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padua, 35122 Padua, Italy; (U.C.); (E.G.); (G.Z.)
| | - Mario Strazzabosco
- Liver Center, Digestive Disease Section, Department of Internal Medicine, Yale University, New Haven, CT 208056, USA;
| | - Paolo Simioni
- Department of Medicine (DIMED), University of Padua, 35122 Padua, Italy;
- General Internal Medicine Unit, Padua University-Hospital, 35122 Padua, Italy
| | - Erica Villa
- Gastroenterology Unit, Department of Medical Specialties, University of Modena & Reggio Emilia and Modena University-Hospital, 41124 Modena, Italy;
- Correspondence: (E.V.); (L.F.); Tel.: +39-059-422-5308 (E.V.); +39-049-821-3131 (L.F.); Fax: +39-059-422-4424 (E.V.); +39-049-827-2355 (L.F.)
| | - Luca Fabris
- Department of Molecular Medicine (DMM), University of Padua, 35122 Padua, Italy;
- Liver Center, Digestive Disease Section, Department of Internal Medicine, Yale University, New Haven, CT 208056, USA;
- General Internal Medicine Unit, Padua University-Hospital, 35122 Padua, Italy
- Correspondence: (E.V.); (L.F.); Tel.: +39-059-422-5308 (E.V.); +39-049-821-3131 (L.F.); Fax: +39-059-422-4424 (E.V.); +39-049-827-2355 (L.F.)
| |
Collapse
|
4
|
Halin Bergström S, Lundholm M, Nordstrand A, Bergh A. Rat prostate tumors induce DNA synthesis in remote organs. Sci Rep 2022; 12:7908. [PMID: 35551231 PMCID: PMC9098422 DOI: 10.1038/s41598-022-12131-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 05/05/2022] [Indexed: 11/09/2022] Open
Abstract
Advanced cancers induce systemic responses. However, if such systemic changes occur already when aggressive tumors are small, have not been thoroughly characterized. Here, we examined how localized prostate cancers of different sizes and metastatic potential affected DNA synthesis in the rest of the prostate and in various remote organs. Non-metastatic Dunning R-3327 G (G) tumor cells, metastatic MatLyLu (MLL) tumor cells, or vehicle were injected into the prostate of immunocompetent rats. All animals received daily injections of Bromodeoxyuridine (BrdU), to label cells/daughter cells with active DNA synthesis. Equal sized G- and MLL-tumors, similarly increased BrdU-labeling in the prostate, lymph nodes and liver compared to tumor-free controls. Prior to metastasis, MLL-tumors also increased BrdU-labeling in bone marrow and lungs compared to animals with G-tumors or controls. In animals with MLL-tumors, BrdU-labeling in prostate, lungs, brown adipose tissue and skeletal muscles increased in a tumor-size-dependent way. Furthermore, MLL-tumors induced increased signs of DNA damage (γH2AX staining) and accumulation of CD68 + macrophages in the lungs. In conclusion, small localized prostate cancers increased DNA synthesis in several remote tissues in a tumor type- and size-dependent way. This may suggest the possibility for early diagnosis of aggressive prostate cancer by examining tumor-induced effects in other tissues.
Collapse
Affiliation(s)
- Sofia Halin Bergström
- Department of Medical Biosciences, Pathology, Umeå University, Building 6M, second floor, 901 87, Umeå, Sweden.
| | - Marie Lundholm
- Department of Medical Biosciences, Pathology, Umeå University, Building 6M, second floor, 901 87, Umeå, Sweden
| | - Annika Nordstrand
- Department of Medical Biosciences, Pathology, Umeå University, Building 6M, second floor, 901 87, Umeå, Sweden
| | - Anders Bergh
- Department of Medical Biosciences, Pathology, Umeå University, Building 6M, second floor, 901 87, Umeå, Sweden
| |
Collapse
|
5
|
Morale MG, Tamura RE, Rubio IGS. Metformin and Cancer Hallmarks: Molecular Mechanisms in Thyroid, Prostate and Head and Neck Cancer Models. Biomolecules 2022; 12:357. [PMID: 35327549 PMCID: PMC8945547 DOI: 10.3390/biom12030357] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 02/01/2023] Open
Abstract
Metformin is the most used drug for type 2 diabetes (T2DM). Its antitumor activity has been described by clinical studies showing reduced risk of cancer development in T2DM patients, as well as management of T2DM compared with those receiving other glucose-lowering drugs. Metformin has a plethora of molecular actions in cancer cells. This review focused on in vitro data on the action mechanisms of metformin on thyroid, prostate and head and neck cancer. AMPK activation regulating specific downstream targets is a constant antineoplastic activity in different types of cancer; however, AMPK-independent mechanisms are also relevant. In vitro evidence makes it clear that depending on the type of tumor, metformin has different actions; its effects may be modulated by different cell conditions (for instance, presence of HPV infection), or it may regulate tissue-specific factors, such as the Na+/I- symporter (NIS) and androgen receptors. The hallmarks of cancer are a set of functional features acquired by the cell during malignant development. In vitro studies show that metformin regulates almost all the hallmarks of cancer. Interestingly, metformin is one of these therapeutic agents with the potential to synergize with other chemotherapeutic agents, with low cost, low side effects and high positive consequences. Some questions are still challenging: Are metformin in vitro data able to translate from bench to bedside? Does metformin affect drug resistance? Can metformin be used as a generic anticancer drug for all types of tumors? Which are the specific actions of metformin on the peculiarities of each type of cancer? Several clinical trials are in progress or have been concluded for repurposing metformin as an anticancer drug. The continuous efforts in the field and future in vitro studies will be essential to corroborate clinical trials results and to elucidate the raised questions.
Collapse
Affiliation(s)
- Mirian Galliote Morale
- Department of Biological Sciences, Federal University of São Paulo, Diadema, Rua Pedro de Toledo 669, 11° Andar, São Paulo 04039-032, Brazil; (M.G.M.); (R.E.T.)
- Laboratory of Cancer Molecular Biology, Federal University of São Paulo, Rua Pedro de Toledo 669, 11° Andar, São Paulo 04039-032, Brazil
| | - Rodrigo Esaki Tamura
- Department of Biological Sciences, Federal University of São Paulo, Diadema, Rua Pedro de Toledo 669, 11° Andar, São Paulo 04039-032, Brazil; (M.G.M.); (R.E.T.)
- Laboratory of Cancer Molecular Biology, Federal University of São Paulo, Rua Pedro de Toledo 669, 11° Andar, São Paulo 04039-032, Brazil
| | - Ileana Gabriela Sanchez Rubio
- Department of Biological Sciences, Federal University of São Paulo, Diadema, Rua Pedro de Toledo 669, 11° Andar, São Paulo 04039-032, Brazil; (M.G.M.); (R.E.T.)
- Laboratory of Cancer Molecular Biology, Federal University of São Paulo, Rua Pedro de Toledo 669, 11° Andar, São Paulo 04039-032, Brazil
- Thyroid Molecular Sciences Laboratory, Federal University of São Paulo, Rua Pedro de Toledo 669, 11° Andar, São Paulo 04039-032, Brazil
| |
Collapse
|
6
|
Carpino G, Cardinale V, Di Giamberardino A, Overi D, Donsante S, Colasanti T, Amato G, Mennini G, Franchitto M, Conti F, Rossi M, Riminucci M, Gaudio E, Alvaro D, Mancone C. Thrombospondin 1 and 2 along with PEDF inhibit angiogenesis and promote lymphangiogenesis in intrahepatic cholangiocarcinoma. J Hepatol 2021; 75:1377-1386. [PMID: 34329660 DOI: 10.1016/j.jhep.2021.07.016] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 07/13/2021] [Accepted: 07/15/2021] [Indexed: 12/27/2022]
Abstract
BACKGROUND & AIMS The microenvironment of intrahepatic cholangiocarcinoma (iCCA) is hypovascularized, with an extensive lymphatic network. This leads to rapid cancer spread into regional lymph nodes and the liver parenchyma, precluding curative treatments. Herein, we investigated which factors released in the iCCA stroma drive the inhibition of angiogenesis and promote lymphangiogenesis. METHODS Quantitative proteomics was performed on extracellular fluid (ECF) proteins extracted both from cancerous and non-cancerous tissues (NCT) of patients with iCCA. Computational biology was applied on a proteomic dataset to identify proteins involved in the regulation of vessel formation. Endothelial cells incubated with ECF from either iCCA or NCT specimens were used to assess the role of candidate proteins in 3D vascular assembly, cell migration, proliferation and viability. Angiogenesis and lymphangiogenesis were further investigated in vivo by a heterotopic transplantation of bone marrow stromal cells, along with endothelial cells in SCID/beige mice. RESULTS Functional analysis of upregulated proteins in iCCA unveils a soluble angio-inhibitory milieu made up of thrombospondin (THBS)1, THBS2 and pigment epithelium-derived factor (PEDF). iCCA ECF was able to inhibit in vitro vessel morphogenesis and viability. Antibodies blocking THBS1, THBS2 and PEDF restored tube formation and endothelial cell viability to levels observed in NCT ECF. Moreover, in transplanted mice, the inhibition of blood vessel formation, the de novo generation of the lymphatic network and the dissemination of iCCA cells in lymph nodes were shown to depend on THBS1, THBS2 and PEDF expression. CONCLUSIONS THBS1, THBS2 and PEDF reduce blood vessel formation and promote tumor-associated lymphangiogenesis in iCCA. Our results identify new potential targets for interventions to counteract the dissemination process in iCCA. LAY SUMMARY Intrahepatic cholangiocarcinoma is a highly aggressive cancer arising from epithelial cells lining the biliary tree, characterized by dissemination into the liver parenchyma via lymphatic vessels. Herein, we show that the proteins THBS1, THBS2 and PEDF, once released in the tumor microenvironment, inhibit vascular growth, while promoting cancer-associated lymphangiogenesis. Therefore, targeting THBS1, THBS2 and PEDF may be a promising strategy to reduce cancer-associated lymphangiogenesis and counteract the invasiveness of intrahepatic cholangiocarcinoma.
Collapse
Affiliation(s)
- Guido Carpino
- Division of Health Sciences, Department of Movement, Human and Health Sciences, University of Rome "Foro Italico", Rome, Italy
| | - Vincenzo Cardinale
- Department of Medico-Surgical Sciences and Biotechnologies, Polo Pontino, Sapienza University of Rome, Rome, Italy
| | | | - Diletta Overi
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Samantha Donsante
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Tania Colasanti
- Rheumatology Unit, Department of Clinical Internal, Anesthetic and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Gaia Amato
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Gianluca Mennini
- General Surgery and Organ Transplantation Unit, Department of General Surgery and Surgical Specialties P. Stefanini, Sapienza University of Rome, Rome, Italy
| | - Matteo Franchitto
- Department of Medical-Surgical Sciences and Translation Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Fabrizio Conti
- Rheumatology Unit, Department of Clinical Internal, Anesthetic and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Massimo Rossi
- General Surgery and Organ Transplantation Unit, Department of General Surgery and Surgical Specialties P. Stefanini, Sapienza University of Rome, Rome, Italy
| | - Mara Riminucci
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Eugenio Gaudio
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Domenico Alvaro
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Carmine Mancone
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
7
|
Abooshahab R, Al-Salami H, Dass CR. The increasing role of pigment epithelium-derived factor in metastasis: from biological importance to a promising target. Biochem Pharmacol 2021; 193:114787. [PMID: 34571004 DOI: 10.1016/j.bcp.2021.114787] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/22/2021] [Accepted: 09/22/2021] [Indexed: 12/17/2022]
Abstract
Pigment epithelium-derived factor (PEDF) is a non-inhibitory member of the serpin (serine protease inhibitor) family and is a well-known potent anti-tumor factor in a variety of cancers. It has been ascertained that PEDF regulates multiple metastatic processes through various plausible mechanisms, including inhibiting angiogenesis, inducing apoptosis, stimulating extracellular matrix (ECM) degradation, and suppressing the epithelial-to-mesenchymal transition (EMT) process. Although PEDF has been recognized as an anti-metastatic marker in most studies, its role remains controversial with conflicting reports of PEDF as a metastatic marker. The emerging insights into the mechanism(s) of PEDF in tumor progression and its therapeutic effects are discussed systematically in this review, aiming to improve our understanding in the context of metastasis and drug development.
Collapse
Affiliation(s)
- Raziyeh Abooshahab
- Curtin Medical School, Curtin University, Bentley 6102, Australia; Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hani Al-Salami
- Curtin Medical School, Curtin University, Bentley 6102, Australia; Curtin Health Innovation Research Institute, Bentley 6102, Australia
| | - Crispin R Dass
- Curtin Medical School, Curtin University, Bentley 6102, Australia; Curtin Health Innovation Research Institute, Bentley 6102, Australia.
| |
Collapse
|
8
|
Koc M, Wald M, Varaliová Z, Ondrůjová B, Čížková T, Brychta M, Kračmerová J, Beranová L, Pala J, Šrámková V, Šiklová M, Gojda J, Rossmeislová L. Lymphedema alters lipolytic, lipogenic, immune and angiogenic properties of adipose tissue: a hypothesis-generating study in breast cancer survivors. Sci Rep 2021; 11:8171. [PMID: 33854130 PMCID: PMC8046998 DOI: 10.1038/s41598-021-87494-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 03/30/2021] [Indexed: 12/26/2022] Open
Abstract
Later stages of secondary lymphedema are associated with the massive deposition of adipose tissue (AT). The factors driving lymphedema-associated AT (LAT) expansion in humans remain rather elusive. We hypothesized that LAT expansion could be based on alterations of metabolic, adipogenic, immune and/or angiogenic qualities of AT. AT samples were acquired from upper limbs of 11 women with unilateral breast cancer-related lymphedema and 11 healthy women without lymphedema. Additional control group of 11 female breast cancer survivors without lymphedema was used to assess systemic effects of lymphedema. AT was analysed for adipocyte size, lipolysis, angiogenesis, secretion of cytokines, immune and stem cell content and mRNA gene expression. Further, adipose precursors were isolated and tested for their proliferative and adipogenic capacity. The effect of undrained LAT- derived fluid on adipogenesis was also examined. Lymphedema did not have apparent systemic effect on metabolism and cytokine levels, but it was linked with higher lymphocyte numbers and altered levels of several miRNAs in blood. LAT showed higher basal lipolysis, (lymph)angiogenic capacity and secretion of inflammatory cytokines when compared to healthy AT. LAT contained more activated CD4+ T lymphocytes than healthy AT. mRNA levels of (lymph)angiogenic markers were deregulated in LAT and correlated with markers of lipolysis. In vitro, adipose cells derived from LAT did not differ in their proliferative, adipogenic, lipogenic and lipolytic potential from cells derived from healthy AT. Nevertheless, exposition of preadipocytes to LAT-derived fluid improved their adipogenic conversion when compared with the effect of serum. This study presents results of first complex analysis of LAT from upper limb of breast cancer survivors. Identified LAT alterations indicate a possible link between (lymph)angiogenesis and lipolysis. In addition, our in vitro results imply that AT expansion in lymphedema could be driven partially by exposition of adipose precursors to undrained LAT-derived fluid.
Collapse
Affiliation(s)
- Michal Koc
- Department of Pathophysiology, Centre for Research On Nutrition, Metabolism and Diabetes, Third Faculty of Medicine, Charles University, Ruská 87, 100 00, Prague 10, Czech Republic
| | - Martin Wald
- Department of Surgery, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague 5, Czech Republic
| | - Zuzana Varaliová
- Department of Pathophysiology, Centre for Research On Nutrition, Metabolism and Diabetes, Third Faculty of Medicine, Charles University, Ruská 87, 100 00, Prague 10, Czech Republic
| | - Barbora Ondrůjová
- Department of Pathophysiology, Centre for Research On Nutrition, Metabolism and Diabetes, Third Faculty of Medicine, Charles University, Ruská 87, 100 00, Prague 10, Czech Republic
| | - Terezie Čížková
- Department of Pathophysiology, Centre for Research On Nutrition, Metabolism and Diabetes, Third Faculty of Medicine, Charles University, Ruská 87, 100 00, Prague 10, Czech Republic
| | - Milan Brychta
- Department of Radiotherapy and Oncology, Kralovske Vinohrady University Hospital, Prague 10, Czech Republic
| | - Jana Kračmerová
- Department of Pathophysiology, Centre for Research On Nutrition, Metabolism and Diabetes, Third Faculty of Medicine, Charles University, Ruská 87, 100 00, Prague 10, Czech Republic
| | - Lenka Beranová
- Department of Pathophysiology, Centre for Research On Nutrition, Metabolism and Diabetes, Third Faculty of Medicine, Charles University, Ruská 87, 100 00, Prague 10, Czech Republic
| | - Jan Pala
- Department of Pathophysiology, Centre for Research On Nutrition, Metabolism and Diabetes, Third Faculty of Medicine, Charles University, Ruská 87, 100 00, Prague 10, Czech Republic
| | - Veronika Šrámková
- Department of Pathophysiology, Centre for Research On Nutrition, Metabolism and Diabetes, Third Faculty of Medicine, Charles University, Ruská 87, 100 00, Prague 10, Czech Republic.,Franco-Czech Laboratory for Clinical Research on Obesity, Third Faculty of Medicine, Prague 10, Czech Republic
| | - Michaela Šiklová
- Department of Pathophysiology, Centre for Research On Nutrition, Metabolism and Diabetes, Third Faculty of Medicine, Charles University, Ruská 87, 100 00, Prague 10, Czech Republic.,Franco-Czech Laboratory for Clinical Research on Obesity, Third Faculty of Medicine, Prague 10, Czech Republic
| | - Jan Gojda
- Franco-Czech Laboratory for Clinical Research on Obesity, Third Faculty of Medicine, Prague 10, Czech Republic.,Second Internal Medicine Department, Kralovske Vinohrady University Hospital, Prague 10, Czech Republic
| | - Lenka Rossmeislová
- Department of Pathophysiology, Centre for Research On Nutrition, Metabolism and Diabetes, Third Faculty of Medicine, Charles University, Ruská 87, 100 00, Prague 10, Czech Republic. .,Franco-Czech Laboratory for Clinical Research on Obesity, Third Faculty of Medicine, Prague 10, Czech Republic.
| |
Collapse
|
9
|
Lipid Metabolism in Tumor-Associated Fibroblasts. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1316:117-131. [PMID: 33740247 DOI: 10.1007/978-981-33-6785-2_8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Tumor- or cancer-associated fibroblasts (TAFs), one of the most abundant stromal cell types in various carcinomas, consist of a heterogeneous cell population. Typically, TAFs are assigned with pro-tumor activities to promote tumor growth and progression. One of the key features of solid tumors is the metabolic reprogramming that induces alterations of bioenergetics and biosynthesis in both tumor cells and TAFs. Therefore, this review emphasizes TAFs lipid metabolism related to both TAFs differentiation process and TAFs crosstalk with cancer cells. We hope that this review will help understand lipid metabolism in tumor microenvironment, and support the rational design of metabolism-based approaches to improve the efficacy of cancer therapy.
Collapse
|
10
|
Brook N, Brook E, Dass CR, Chan A, Dharmarajan A. Pigment Epithelium-Derived Factor and Sex Hormone-Responsive Cancers. Cancers (Basel) 2020; 12:cancers12113483. [PMID: 33238558 PMCID: PMC7700359 DOI: 10.3390/cancers12113483] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 12/24/2022] Open
Abstract
Oestrogens and androgens play important roles in normal and cancerous tissue and have been shown to negatively regulate pigment epithelium-derived factor (PEDF) expression in sex hormone-responsive tumours. PEDF suppresses tumour growth and its downregulation by oestrogen is implicated in tumorigenesis, metastasis, and progression. PEDF expression is reduced in cancerous tissue of the prostate, breast, ovary, and endometrium compared to their normal tissue counterparts, with a link between PEDF downregulation and sex hormone signalling observed in pre-clinical studies. PEDF reduces growth and metastasis of tumour cells by promoting apoptosis, inhibiting angiogenesis, increasing adhesion, and reducing migration. PEDF may also prevent treatment resistance in some cancers by downregulating oestrogen receptor signalling. By interacting with components of the tumour microenvironment, PEDF counteracts the proliferative and immunosuppressive effects of oestrogens, to ultimately reduce tumorigenesis and metastasis. In this review, we focus on sex hormone regulation of PEDF's anti-tumour action in sex hormone-responsive tumours.
Collapse
Affiliation(s)
- Naomi Brook
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, WA 6102, Australia; (N.B.); (E.B.)
- Curtin Health Innovation Research Institute, Bentley, WA 6102, Australia
| | - Emily Brook
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, WA 6102, Australia; (N.B.); (E.B.)
- Curtin Health Innovation Research Institute, Bentley, WA 6102, Australia
| | - Crispin R. Dass
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, WA 6102, Australia; (N.B.); (E.B.)
- Curtin Health Innovation Research Institute, Bentley, WA 6102, Australia
- Correspondence: (C.R.D.); (A.D.); Tel.: +61-8-9266-1489 (C.R.D.)
| | - Arlene Chan
- School of Medicine, Curtin University, Bentley, WA 6102, Australia;
- Breast Cancer Research Centre-Western Australia, Hollywood Private Hospital, Nedlands, WA 6009, Australia
| | - Arun Dharmarajan
- Department of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai 600116, India
- Correspondence: (C.R.D.); (A.D.); Tel.: +61-8-9266-1489 (C.R.D.)
| |
Collapse
|
11
|
Yamagishi SI, Koga Y, Sotokawauchi A, Hashizume N, Fukahori S, Matsui T, Yagi M. Therapeutic Potential of Pigment Epithelium-derived Factor in Cancer. Curr Pharm Des 2020; 25:313-324. [PMID: 30892156 DOI: 10.2174/1381612825666190319112106] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 03/13/2019] [Indexed: 12/11/2022]
Abstract
Pigment epithelium-derived factor (PEDF) is one of the serine protease inhibitors with multifunctional properties, which is produced by various types of organs and tissues. There is an accumulating body of evidence that PEDF plays an important role in the maintenance of tissue homeostasis. Indeed, PEDF not only works as an endogenous inhibitor of angiogenesis, but also suppresses oxidative stress, inflammatory and thrombotic reactions in cell culture systems, animal models, and humans. Furthermore, we, along with others, have found that PEDF inhibits proliferation of, and induces apoptotic cell death in, numerous kinds of tumors. In addition, circulating as well as tumor expression levels of PEDF have been inversely associated with tumor growth and metastasis. These observations suggest that supplementation of PEDF proteins and/or enhancement of endogenous PEDF expression could be a novel therapeutic strategy for the treatment of cancer. Therefore, in this paper, we review the effects of PEDF on diverse types of cancer, and discuss its therapeutic perspectives.
Collapse
Affiliation(s)
- Sho-Ichi Yamagishi
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume 830-0011, Japan
| | - Yoshinori Koga
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume 830-0011, Japan.,Department of Pediatric Surgery, Kurume University School of Medicine, Kurume 830-0011, Japan
| | - Ami Sotokawauchi
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume 830-0011, Japan
| | - Naoki Hashizume
- Department of Pediatric Surgery, Kurume University School of Medicine, Kurume 830-0011, Japan
| | - Suguru Fukahori
- Department of Pediatric Surgery, Kurume University School of Medicine, Kurume 830-0011, Japan
| | - Takanori Matsui
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume 830-0011, Japan
| | - Minoru Yagi
- Department of Pediatric Surgery, Kurume University School of Medicine, Kurume 830-0011, Japan
| |
Collapse
|
12
|
Nardi F, Fitchev P, Brooks KM, Franco OE, Cheng K, Hayward SW, Welte MA, Crawford SE. Lipid droplet velocity is a microenvironmental sensor of aggressive tumors regulated by V-ATPase and PEDF. J Transl Med 2019; 99:1822-1834. [PMID: 31409893 PMCID: PMC7289525 DOI: 10.1038/s41374-019-0296-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 06/04/2019] [Accepted: 06/05/2019] [Indexed: 12/19/2022] Open
Abstract
Lipid droplets (LDs) utilize microtubules (MTs) to participate in intracellular trafficking of cargo proteins. Cancer cells accumulate LDs and acidify their tumor microenvironment (TME) by increasing the proton pump V-ATPase. However, it is not known whether these two metabolic changes are mechanistically related or influence LD movement. We postulated that LD density and velocity are progressively increased with tumor aggressiveness and are dependent on V-ATPase and the lipolysis regulator pigment epithelium-derived factor (PEDF). LD density was assessed in human prostate cancer (PCa) specimens across Gleason scores (GS) 6-8. LD distribution and velocity were analyzed in low and highly aggressive tumors using live-cell imaging and in cells exposed to low pH and/or treated with V-ATPase inhibitors. The MT network was disrupted and analyzed by α-tubulin staining. LD density positively correlated with advancing GS in human tumors. Acidification promoted peripheral localization and clustering of LDs. Highly aggressive prostate, breast, and pancreatic cell lines had significantly higher maximum LD velocity (LDVmax) than less aggressive and benign cells. LDVmax was MT-dependent and suppressed by blocking V-ATPase directly or indirectly with PEDF. Upon lowering pH, LDs moved to the cell periphery and carried metalloproteinases. These results suggest that acidification of the TME can alter intracellular LD movement and augment velocity in cancer. Restoration of PEDF or blockade of V-ATPase can normalize LD distribution and decrease velocity. This study identifies V-ATPase and PEDF as new modulators of LD trafficking in the cancer microenvironment.
Collapse
Affiliation(s)
- Francesca Nardi
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL 60201
| | - Philip Fitchev
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL 60201
| | - Kyrsten M. Brooks
- Department of Pathology, Saint Louis University School of Medicine, 1402 South Grand Blvd., Saint Louis, MO 63104
| | - Omar E. Franco
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL 60201
| | - Kevin Cheng
- Department of Pathology, Saint Louis University School of Medicine, 1402 South Grand Blvd., Saint Louis, MO 63104
| | - Simon W. Hayward
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL 60201
| | - Michael A. Welte
- Department of Biology, University of Rochester, RC Box 270211, Rochester, NY 14627
| | - Susan E. Crawford
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL 60201,Department of Pathology, Saint Louis University School of Medicine, 1402 South Grand Blvd., Saint Louis, MO 63104
| |
Collapse
|
13
|
Sag D, Ayyildiz ZO, Gunalp S, Wingender G. The Role of TRAIL/DRs in the Modulation of Immune Cells and Responses. Cancers (Basel) 2019; 11:cancers11101469. [PMID: 31574961 PMCID: PMC6826877 DOI: 10.3390/cancers11101469] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/09/2019] [Accepted: 09/20/2019] [Indexed: 12/26/2022] Open
Abstract
Expression of TRAIL (tumor necrosis factor–related apoptosis–inducing ligand) by immune cells can lead to the induction of apoptosis in tumor cells. However, it becomes increasingly clear that the interaction of TRAIL and its death receptors (DRs) can also directly impact immune cells and influence immune responses. Here, we review what is known about the role of TRAIL/DRs in immune cells and immune responses in general and in the tumor microenvironment in particular.
Collapse
Affiliation(s)
- Duygu Sag
- Izmir Biomedicine and Genome Center (IBG), 35340 Balcova/Izmir, Turkey.
- Department of Medical Biology, Faculty of Medicine, Dokuz Eylul University, 35340 Balcova/Izmir, Turkey.
- Department of Genome Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340 Balcova/Izmir, Turkey.
| | - Zeynep Ozge Ayyildiz
- Department of Genome Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340 Balcova/Izmir, Turkey.
| | - Sinem Gunalp
- Department of Genome Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340 Balcova/Izmir, Turkey.
| | - Gerhard Wingender
- Izmir Biomedicine and Genome Center (IBG), 35340 Balcova/Izmir, Turkey.
- Department of Biomedicine and Health Technologies, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340 Balcova/Izmir, Turkey.
| |
Collapse
|
14
|
Bishehsari F, Zhang L, Barlass U, Preite N, Turturro S, Najor MS, Shetuni BB, Zayas JP, Mahdavinia M, Abukhdeir AM, Keshavarzian A. KRAS mutation and epithelial-macrophage interplay in pancreatic neoplastic transformation. Int J Cancer 2018; 143:1994-2007. [PMID: 29756386 PMCID: PMC6128758 DOI: 10.1002/ijc.31592] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 02/27/2018] [Accepted: 04/11/2018] [Indexed: 01/01/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDA) is characterized by epithelial mutations in KRAS and prominent tumor-associated inflammation, including macrophage infiltration. But knowledge of early interactions between neoplastic epithelium and macrophages in PDA carcinogenesis is limited. Using a pancreatic organoid model, we found that the expression of mutant KRAS in organoids increased (i) ductal to acinar gene expression ratios, (ii) epithelial cells proliferation and (iii) colony formation capacity in vitro, and endowed pancreatic cells with the ability to generate neoplastic tumors in vivo. KRAS mutations induced a protumorigenic phenotype in macrophages. Altered macrophages decreased epithelial pigment epithelial derived factor (PEDF) expression and induced a cancerous phenotype. We validated our findings using annotated patient samples from The Cancer Genome Atlas (TCGA) and in our human PDA specimens. Epithelium-macrophage cross-talk occurs early in pancreatic carcinogenesis where KRAS directly induces cancer-related phenotypes in epithelium, and also promotes a protumorigenic phenotype in macrophages, in turn augmenting neoplastic growth.
Collapse
Affiliation(s)
- Faraz Bishehsari
- Department of Medicine, Division of Gastroenterology, Rush University Medical Center, Chicago, IL 60612
| | - Lijuan Zhang
- Department of Medicine, Division of Gastroenterology, Rush University Medical Center, Chicago, IL 60612
| | - Usman Barlass
- Department of Medicine, Rush University Medical Center, Chicago, Illinois
| | - Nailliw Preite
- Department of Medicine, Division of Gastroenterology, Rush University Medical Center, Chicago, IL 60612
| | - Sanja Turturro
- Department of Medicine, Division of Hematology, Oncology, and Cell Therapy, Rush University Medical Center, Chicago, IL 60612
| | - Matthew S. Najor
- Department of Medicine, Division of Hematology, Oncology, and Cell Therapy, Rush University Medical Center, Chicago, IL 60612
| | | | - Janet P Zayas
- Department of Medicine, Rush University Medical Center, Chicago, Illinois
| | - Mahboobeh Mahdavinia
- Department of Medicine, Division of Allergy-Immunology, Rush University Medical Center, Chicago, IL 60612
| | - Abde M. Abukhdeir
- Department of Medicine, Division of Hematology, Oncology, and Cell Therapy, Rush University Medical Center, Chicago, IL 60612
| | - Ali Keshavarzian
- Department of Medicine, Division of Gastroenterology, Rush University Medical Center, Chicago, IL 60612
| |
Collapse
|
15
|
Nardi F, Fitchev P, Franco OE, Ivanisevic J, Scheibler A, Hayward SW, Brendler CB, Welte MA, Crawford SE. PEDF regulates plasticity of a novel lipid-MTOC axis in prostate cancer-associated fibroblasts. J Cell Sci 2018; 131:jcs.213579. [PMID: 29792311 DOI: 10.1242/jcs.213579] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 05/10/2018] [Indexed: 12/27/2022] Open
Abstract
Prostate tumors make metabolic adaptations to ensure adequate energy and amplify cell cycle regulators, such as centrosomes, to sustain their proliferative capacity. It is not known whether cancer-associated fibroblasts (CAFs) undergo metabolic re-programming. We postulated that CAFs augment lipid storage and amplify centrosomal or non-centrosomal microtubule-organizing centers (MTOCs) through a pigment epithelium-derived factor (PEDF)-dependent lipid-MTOC signaling axis. Primary human normal prostate fibroblasts (NFs) and CAFs were evaluated for lipid content, triacylglycerol-regulating proteins, MTOC number and distribution. CAFs were found to store more neutral lipids than NFs. Adipose triglyceride lipase (ATGL) and PEDF were strongly expressed in NFs, whereas CAFs had minimal to undetectable levels of PEDF or ATGL protein. At baseline, CAFs demonstrated MTOC amplification when compared to 1-2 perinuclear MTOCs consistently observed in NFs. Treatment with PEDF or blockade of lipogenesis suppressed lipid content and MTOC number. In summary, our data support that CAFs have acquired a tumor-like phenotype by re-programming lipid metabolism and amplifying MTOCs. Normalization of MTOCs by restoring PEDF or by blocking lipogenesis highlights a previously unrecognized plasticity in centrosomes, which is regulated through a new lipid-MTOC axis.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Francesca Nardi
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL 60201, United States
| | - Philip Fitchev
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL 60201, United States
| | - Omar E Franco
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL 60201, United States
| | - Jelena Ivanisevic
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL 60201, United States
| | - Adrian Scheibler
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL 60201, United States
| | - Simon W Hayward
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL 60201, United States
| | - Charles B Brendler
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL 60201, United States
| | - Michael A Welte
- Department of Biology, University of Rochester, Rochester, NY 14627, United States
| | - Susan E Crawford
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL 60201, United States
| |
Collapse
|
16
|
Principe DR, DeCant B, Diaz AM, Mangan RJ, Hwang R, Lowy A, Shetuni BB, Sreekumar BK, Chung C, Bentrem DJ, Munshi HG, Jung B, Grippo PJ, Bishehsari F. PEDF inhibits pancreatic tumorigenesis by attenuating the fibro-inflammatory reaction. Oncotarget 2017; 7:28218-34. [PMID: 27058416 PMCID: PMC5053722 DOI: 10.18632/oncotarget.8587] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 03/17/2016] [Indexed: 12/19/2022] Open
Abstract
Pancreatic cancer is characterized by a pronounced fibro-inflammatory reaction that has been shown to contribute to cancer progression. Previous reports have demonstrated that pigment epithelium-derived factor (PEDF) has potent tumor suppressive effects in pancreatic cancer, though little is known about the mechanisms by which PEDF limits pancreatic tumorigenesis. We therefore employed human specimens, as well as mouse and in vitro models, to explore the effects of PEDF upon the pancreatic microenvironment. We found that PEDF expression is decreased in human pancreatic cancer samples compared to non-malignant tissue. Furthermore, PEDF-deficient patients displayed increased intratumoral inflammation/fibrosis. In mice, genetic ablation of PEDF increased cerulein-induced inflammation and fibrosis, and similarly enhanced these events in the background of oncogenic KRAS. In vitro, recombinant PEDF neutralized macrophage migration as well as inhibited macrophage-induced proliferation of tumor cells. Additionally, recombinant PEDF suppressed the synthesis of pro-inflammatory/pro-fibrotic cytokines both in vivo and in vitro, and reduced collagen I deposition and TGFβ synthesis by pancreatic stellate cells, consistent with reduced fibrosis. Combined, our results demonstrate that PEDF limits pancreatic cancer progression by attenuating the fibro-inflammatory reaction, and makes restoration of PEDF signaling a potential therapeutic approach to study in pancreatic cancer.
Collapse
Affiliation(s)
| | - Brian DeCant
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Andrew M Diaz
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Riley J Mangan
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Rosa Hwang
- Department of Surgical Oncology, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Andrew Lowy
- Department of Surgery, University of California San Diego, San Diego, CA, USA
| | | | - Bharath K Sreekumar
- Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Chuhan Chung
- Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - David J Bentrem
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Hidayatullah G Munshi
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Barbara Jung
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Paul J Grippo
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Faraz Bishehsari
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
17
|
Flores-Morales A, Iglesias-Gato D. Quantitative Mass Spectrometry-Based Proteomic Profiling for Precision Medicine in Prostate Cancer. Front Oncol 2017; 7:267. [PMID: 29164064 PMCID: PMC5674010 DOI: 10.3389/fonc.2017.00267] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 10/23/2017] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer (PCa) is one of the most frequently diagnosed cancer among men in the western societies. Many PCa patients bear tumors that will not threat their lives if left untreated or if treatment is delayed. Our inability for early identification of these patients has resulted in massive overtreatment. Therefore, there is a great need of finding biomarkers for patient stratification according to prognostic risk; as well as there is a need for novel targets that can allow the development of effective treatments for patients that progress to castration-resistant PCa. Most biomarkers in cancer are proteins, including the widely-used prostate-specific antigen (PSA). Recent developments in mass spectrometry allow the identification and quantification of thousands of proteins and posttranslational modifications from small amounts of biological material, including formalin-fixed paraffin-embedded tissues, and biological fluids. Novel diagnostic and prognostic biomarkers have been identified in tissue, blood, urine, and seminal plasma of PCa patients, and new insights in the ethology and progression of this disease have been achieved using this technology. In this review, we summarize these findings and discuss the potential of this technology to pave the way toward the clinical implementation of precision medicine in PCa.
Collapse
Affiliation(s)
- Amilcar Flores-Morales
- Faculty of Health and Medical Sciences, Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark.,Danish Cancer Society Research Center, Danish Cancer Society, Copenhagen, Denmark
| | - Diego Iglesias-Gato
- Faculty of Health and Medical Sciences, Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark.,Danish Cancer Society Research Center, Danish Cancer Society, Copenhagen, Denmark
| |
Collapse
|
18
|
Martinez-Marin D, Jarvis C, Nelius T, de Riese W, Volpert OV, Filleur S. PEDF increases the tumoricidal activity of macrophages towards prostate cancer cells in vitro. PLoS One 2017; 12:e0174968. [PMID: 28403150 PMCID: PMC5389654 DOI: 10.1371/journal.pone.0174968] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 03/19/2017] [Indexed: 12/23/2022] Open
Abstract
Background Although inflammation and prostate cancer (PCa) have been linked, the molecular interactions between macrophages and PCa cells are poorly explored. Pigment Epithelium-Derived Factor (PEDF) is an anti-angiogenic and anti-tumor factor. We previously showed that PEDF induces macrophages recruitment in vitro, correlates with macrophages density in human prostate, and stimulates macrophages polarization towards the classically activated pathway. Here, we demonstrate that PEDF modulates the interaction between macrophages and PCa cells through a bidirectional signalling leading to tumor cell apoptosis and phagocytosis. Methods RAW 264.7 and THP-1 cells, and BMDMs were grown in vitro as mono- or co-cultures with PC3 or CL1 tumor cells. The effects of PEDF and its derived P18 peptide were measured on macrophages differentiation, migration, and superoxide production, and tumor cell apoptosis and phagocytosis. PEDF receptors (ATP5B, PNPLA2, and LRP6) and CD47 mRNA and protein expression were quantified in macrophages and tumor cells by quantitative RT-PCR, western blot, immunofluorescence and flow cytometry. Results We found that PEDF induced the migration of macrophages towards tumor 3D spheroids and 2D cultures. In co-culture, PEDF increased PCa cells phagocytosis through an indirect apoptosis-dependent mechanism. Moreover, PEDF stimulated the production of superoxide by macrophages. Conditioned media from macrophages exposed to PEDF induced tumor cells apoptosis in contrast to control conditioned media suggesting that ROS may be involved in tumor cells apoptosis. ATP5B and PNPLA2 PEDF receptors on macrophages and CD47 on tumor cells were respectively up- and down-regulated by PEDF. As PEDF, blocking CD47 induced phagocytosis. Inhibiting ATP5B reduced phagocytosis. Inversely, PNPLA2 inhibition blocks differentiation but maintains phagocytosis. CD47-induced phagocytosis was partially reverted by ATP5B inhibition suggesting a complementary action. Similar effects were observed with P18 PEDF-derived peptide. Conclusions These data established that modulating the molecular interactions between macrophages and PCa cells using PEDF may be a promising strategy for PCa treatment.
Collapse
Affiliation(s)
- Dalia Martinez-Marin
- Department of Urology, Texas Tech University-Health Sciences Center, Lubbock, Texas, United States of America
| | - Courtney Jarvis
- Department of Urology, Texas Tech University-Health Sciences Center, Lubbock, Texas, United States of America
- Department of Immunology and Molecular Microbiology, Texas Tech University-Health Sciences Center, Lubbock, Texas, United States of America
| | - Thomas Nelius
- Department of Urology, Texas Tech University-Health Sciences Center, Lubbock, Texas, United States of America
| | - Werner de Riese
- Department of Urology, Texas Tech University-Health Sciences Center, Lubbock, Texas, United States of America
| | - Olga V. Volpert
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Stéphanie Filleur
- Department of Urology, Texas Tech University-Health Sciences Center, Lubbock, Texas, United States of America
- Department of Immunology and Molecular Microbiology, Texas Tech University-Health Sciences Center, Lubbock, Texas, United States of America
- * E-mail:
| |
Collapse
|
19
|
Halin Bergström S, Hägglöf C, Thysell E, Bergh A, Wikström P, Lundholm M. Extracellular Vesicles from Metastatic Rat Prostate Tumors Prime the Normal Prostate Tissue to Facilitate Tumor Growth. Sci Rep 2016; 6:31805. [PMID: 27550147 PMCID: PMC4994101 DOI: 10.1038/srep31805] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 07/26/2016] [Indexed: 12/21/2022] Open
Abstract
Accumulating data indicates that tumor-derived extracellular vesicles (EVs) are responsible for tumor-promoting effects. However, if tumor EVs also prepare the tumor-bearing organ for subsequent tumor growth, and if this effect is different in low and high malignant tumors is not thoroughly explored. Here we used orthotopic rat Dunning R-3327 prostate tumors to compare the role of EVs from fast growing and metastatic MatLyLu (MLL) tumors with EVs from more indolent and non-metastatic Dunning G (G) tumors. Prostate tissue pre-conditioned with MLL-EVs in vivo facilitated G tumor establishment compared to G-EVs. MLL-EVs increased prostate epithelial proliferation and macrophage infiltration into the prostate compared to G-EVs. Both types of EVs increased macrophage endocytosis and the mRNA expression of genes associated with M2 polarization in vitro, with MLL-EVs giving the most pronounced effects. MLL-EVs also altered the mRNA expression of growth factors and cytokines in primary rat prostate fibroblasts compared to G-EVs, suggesting fibroblast activation. Our findings propose that EVs from metastatic tumors have the ability to prime the prostate tissue and enhance tumor growth to a higher extent than EVs from non-metastatic tumors. Identifying these differences could lead to novel therapeutic targets and potential prognostic markers for prostate cancer.
Collapse
Affiliation(s)
| | - Christina Hägglöf
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Elin Thysell
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Anders Bergh
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Pernilla Wikström
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Marie Lundholm
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| |
Collapse
|
20
|
Halin Bergström S, Nilsson M, Adamo H, Thysell E, Jernberg E, Stattin P, Widmark A, Wikström P, Bergh A. Extratumoral Heme Oxygenase-1 (HO-1) Expressing Macrophages Likely Promote Primary and Metastatic Prostate Tumor Growth. PLoS One 2016; 11:e0157280. [PMID: 27280718 PMCID: PMC4900522 DOI: 10.1371/journal.pone.0157280] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 05/26/2016] [Indexed: 01/06/2023] Open
Abstract
Aggressive tumors induce tumor-supporting changes in the benign parts of the prostate. One factor that has increased expression outside prostate tumors is hemoxygenase-1 (HO-1). To investigate HO-1 expression in more detail, we analyzed samples of tumor tissue and peritumoral normal prostate tissue from rats carrying cancers with different metastatic capacity, and human prostate cancer tissue samples from primary tumors and bone metastases. In rat prostate tumor samples, immunohistochemistry and quantitative RT-PCR showed that the main site of HO-1 synthesis was HO-1+ macrophages that accumulated in the tumor-bearing organ, and at the tumor-invasive front. Small metastatic tumors were considerably more effective in attracting HO-1+ macrophages than larger non-metastatic ones. In clinical samples, accumulation of HO-1+ macrophages was seen at the tumor invasive front, almost exclusively in high-grade tumors, and it correlated with the presence of bone metastases. HO-1+ macrophages, located at the tumor invasive front, were more abundant in bone metastases than in primary tumors. HO-1 expression in bone metastases was variable, and positively correlated with the expression of macrophage markers but negatively correlated with androgen receptor expression, suggesting that elevated HO-1 could be a marker for a subgroup of bone metastases. Together with another recent observation showing that selective knockout of HO-1 in macrophages reduced prostate tumor growth and metastatic capacity in animals, the results of this study suggest that extratumoral HO-1+ macrophages may have an important role in prostate cancer.
Collapse
Affiliation(s)
- Sofia Halin Bergström
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
- * E-mail:
| | - Maria Nilsson
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Hanibal Adamo
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Elin Thysell
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Emma Jernberg
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Pär Stattin
- Department of Surgical and Perioperative Sciences, Urology, Umeå University, Umeå, Sweden
| | - Anders Widmark
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| | - Pernilla Wikström
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Anders Bergh
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| |
Collapse
|
21
|
Nwani NG, Deguiz ML, Jimenez B, Vinokour E, Dubrovskyi O, Ugolkov A, Mazar AP, Volpert OV. Melanoma Cells Block PEDF Production in Fibroblasts to Induce the Tumor-Promoting Phenotype of Cancer-Associated Fibroblasts. Cancer Res 2016; 76:2265-76. [PMID: 26921338 DOI: 10.1158/0008-5472.can-15-2468] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 02/06/2016] [Indexed: 01/28/2023]
Abstract
Loss of pigment epithelium-derived factor (PEDF, SERPINF1) in cancer cells is associated with poor prognosis and metastasis, but the contribution of stromal PEDF to cancer evolution is poorly understood. Therefore, we investigated the role of fibroblast-derived PEDF in melanoma progression. We demonstrate that normal dermal fibroblasts expressing high PEDF levels attenuated melanoma growth and angiogenesis in vivo, whereas PEDF-depleted fibroblasts exerted tumor-promoting effects. Accordingly, mice with global PEDF knockout were more susceptible to melanoma metastasis. We also demonstrate that normal fibroblasts in close contact with PEDF-null melanoma cells lost PEDF expression and tumor-suppressive properties. Further mechanistic investigations underlying the crosstalk between tumor and stromal cells revealed that melanoma cells produced PDGF-BB and TGFβ, which blocked PEDF production in fibroblasts. Notably, cancer-associated fibroblasts (CAF) isolated from patient-derived tumors expressed markedly low levels of PEDF. Treatment of patient CAF and TGFβ-treated normal fibroblasts with exogenous PEDF decreased the expression of CAF markers and restored PEDF expression. Finally, expression profiling of PEDF-depleted fibroblasts revealed induction of IL8, SERPINB2, hyaluronan synthase-2, and other genes associated with tumor promotion and metastasis. Collectively, our results demonstrate that PEDF maintains tumor-suppressive functions in fibroblasts to prevent CAF conversion and illustrate the mechanisms by which melanoma cells silence stromal PEDF to promote malignancy. Cancer Res; 76(8); 2265-76. ©2016 AACR.
Collapse
Affiliation(s)
- Nkechiyere G Nwani
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Maria L Deguiz
- Department of Biochemistry, Universidad Autónoma de Madrid, Madrid, Spain. Instituto de Investigaciones Biomédicas Alberto Sols CSIC-UAM, Madrid, Spain. Instituto de Investigación I+12, Madrid, Spain
| | - Benilde Jimenez
- Department of Biochemistry, Universidad Autónoma de Madrid, Madrid, Spain. Instituto de Investigaciones Biomédicas Alberto Sols CSIC-UAM, Madrid, Spain. Instituto de Investigación I+12, Madrid, Spain
| | - Elena Vinokour
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Oleksii Dubrovskyi
- Northwestern University Center for Developmental Therapeutics, Evanston, Illinois
| | - Andrey Ugolkov
- Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois
| | - Andrew P Mazar
- Northwestern University Center for Developmental Therapeutics, Evanston, Illinois. Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois. Robert H Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois. Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Olga V Volpert
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, Illinois. Northwestern University Center for Developmental Therapeutics, Evanston, Illinois. Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois. Robert H Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois. Feinberg Cardiovascular Institute at Northwestern University Feinberg School of Medicine, Chicago, Illinois Illinois.
| |
Collapse
|
22
|
Belkacemi L, Zhang SX. Anti-tumor effects of pigment epithelium-derived factor (PEDF): implication for cancer therapy. A mini-review. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2016; 35:4. [PMID: 26746675 PMCID: PMC4706649 DOI: 10.1186/s13046-015-0278-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 12/22/2015] [Indexed: 12/22/2022]
Abstract
Pigment epithelium-derived factor (PEDF) is a secreted glycoprotein and a non-inhibitory member of the serine protease inhibitor (serpin) family. It is widely expressed in human fetal and adult tissues but its expression decreases with age and in malignant tissues. The main anti-cancer activities of PEDF derive from its dual effects, either indirectly on the tumor microenvironment (indirect antitumor action) or directly on the tumor itself (direct antitumor influence). The indirect antitumor activities of PEDF were uncovered from the early findings that it stimulates retinoblastoma cell differentiation and that additionally it possesses anti-angiogenic, anti-tumorigenic and anti-metastatic properties. The mechanisms of its direct antitumor effect, however, have not been fully elucidated. This review highlights recent progress in our understanding of the multifunctional activities of PEDF and, in particular, its anti-cancer signaling mechanisms. Additionally, we discuss the possibility of using novel phosphaplatin compounds that can upregulate PEDF expression as a chemotherapy for cancer treatment.
Collapse
Affiliation(s)
- Louiza Belkacemi
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, 77204, USA.
| | - Shaun Xiaoliu Zhang
- Department of Biology and Biochemistry, University of Houston, Houston, TX, 77204, USA. .,Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, 77204, USA.
| |
Collapse
|
23
|
Adamo HH, Strömvall K, Nilsson M, Halin Bergström S, Bergh A. Adaptive (TINT) Changes in the Tumor Bearing Organ Are Related to Prostate Tumor Size and Aggressiveness. PLoS One 2015; 10:e0141601. [PMID: 26536349 PMCID: PMC4633147 DOI: 10.1371/journal.pone.0141601] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 10/09/2015] [Indexed: 11/18/2022] Open
Abstract
In order to grow, tumors need to induce supportive alterations in the tumor-bearing organ, by us named tumor instructed normal tissue (TINT) changes. We now examined if the nature and magnitude of these responses were related to tumor size and aggressiveness. Three different Dunning rat prostate tumor cells were implanted into the prostate of immune-competent rats; 1) fast growing and metastatic MatLyLu tumor cells 2) fast growing and poorly metastatic AT-1 tumor cells, and 3) slow growing and non-metastatic G tumor cells. All tumor types induced increases in macrophage, mast cell and vascular densities and in vascular cell-proliferation in the tumor-bearing prostate lobe compared to controls. These increases occurred in parallel with tumor growth. The most pronounced and rapid responses were seen in the prostate tissue surrounding MatLyLu tumors. They were, also when small, particularly effective in attracting macrophages and stimulating growth of not only micro-vessels but also small arteries and veins compared to the less aggressive AT-1 and G tumors. The nature and magnitude of tumor-induced changes in the tumor-bearing organ are related to tumor size but also to tumor aggressiveness. These findings, supported by previous observation in patient samples, suggest that one additional way to evaluate prostate tumor aggressiveness could be to monitor its effect on adjacent tissues.
Collapse
Affiliation(s)
- Hanibal Hani Adamo
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Kerstin Strömvall
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Maria Nilsson
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | | | - Anders Bergh
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
- * E-mail:
| |
Collapse
|
24
|
Proapoptotic PEDF functional peptides inhibit prostate tumor growth--a mechanistic study. Biochem Pharmacol 2014; 92:425-37. [PMID: 25261795 DOI: 10.1016/j.bcp.2014.09.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Revised: 09/13/2014] [Accepted: 09/17/2014] [Indexed: 11/22/2022]
Abstract
PEDF inhibits tumor growth via anti-angiogenic activity; however, the direct effect of PEDF on prostate carcinoma and its functional epitope as well as the underlying mechanism regulating the pathway from extracellular receptors to nuclear transcription factors has not been fully elucidated. This study investigates the ability and mechanism by which the functional PEDF peptides PEDF34 and PEDF44 suppress tumor growth. The results showed that death receptor pathway was activated by PEDF34 through up-regulation of FasL and activation of caspase-8 in both xenograft tumor tissues and PC-3 cells. FasL knockdown by siRNA or JNK-p inhibition attenuated apoptosis induced by PEDF34. NF-κB and PPARγ are crucial transcription factors for FasL expression. PEDF34 up-regulated PPARγ but did not affect NF-κB. PEDF34-induced up-regulation of FasL was abolished by siRNA-mediated PPARγ knockdown or using PPARγ inhibitor GW9662, whereas inhibition of NF-κB by the inhibitor PDTC or by siRNA had no effect. Furthermore, activation of JNK is necessary for PEDF34-induced up-regulation of FasL. PEDF34 has stronger hydropathicity and more interactions with laminin receptor than PEDF44. Blocking the laminin receptor abolished the up-regulation of FasL and PPARγ by PEDF34. Moreover, PEDF34 uses a similar mechanism to induce apoptosis in both endothelial and cancer cells. This study provides evidence that PEDF34, not PEDF44, serves as the proapoptotic epitope and exerts proapoptotic activity in both cancer and endothelial cells through activation of the extrinsic death receptor pathway. The dual anti-tumor and anti-angiogenic activities of PEDF34 suggest that it may be a promising agent for the treatment of prostate cancer.
Collapse
|
25
|
Roet KCD, Verhaagen J. Understanding the neural repair-promoting properties of olfactory ensheathing cells. Exp Neurol 2014; 261:594-609. [PMID: 24842489 DOI: 10.1016/j.expneurol.2014.05.007] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 05/02/2014] [Accepted: 05/06/2014] [Indexed: 12/13/2022]
Abstract
Olfactory ensheathing glial cells (OECs) are a specialized type of glia that form a continuously aligned cellular pathway that actively supports unprecedented regeneration of primary olfactory axons from the periphery into the central nervous system. Implantation of OECs stimulates neural repair in experimental models of spinal cord, brain and peripheral nerve injury and delays disease progression in animal models for neurodegenerative diseases like amyotrophic lateral sclerosis. OECs implanted in the injured spinal cord display a plethora of pro-regenerative effects; they promote axonal regeneration, reorganize the glial scar, remyelinate axons, stimulate blood vessel formation, have phagocytic properties and modulate the immune response. Recently genome wide transcriptional profiling and proteomics analysis combined with classical or larger scale "medium-throughput" bioassays have provided novel insights into the molecular mechanism that endow OECs with their pro-regenerative properties. Here we review these studies and show that the gaps that existed in our understanding of the molecular basis of the reparative properties of OECs are narrowing. OECs express functionally connected sets of genes that can be linked to at least 10 distinct processes directly relevant to neural repair. The data indicate that OECs exhibit a range of synergistic cellular activities, including active and passive stimulation of axon regeneration (by secretion of growth factors, axon guidance molecules and basement membrane components) and critical aspects of tissue repair (by structural remodeling and support, modulation of the immune system, enhancement of neurotrophic and antigenic stimuli and by metabolizing toxic macromolecules). Future experimentation will have to further explore the newly acquired knowledge to enhance the therapeutic potential of OECs.
Collapse
Affiliation(s)
- Kasper C D Roet
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105BA Amsterdam, The Netherlands.
| | - Joost Verhaagen
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105BA Amsterdam, The Netherlands; Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, Boelelaan 1085, Amsterdam 1081HV, The Netherlands.
| |
Collapse
|
26
|
Nelius T, Martinez-Marin D, Hirsch J, Miller B, Rinard K, Lopez J, de Riese W, Filleur S. Pigment epithelium-derived factor expression prolongs survival and enhances the cytotoxicity of low-dose chemotherapy in castration-refractory prostate cancer. Cell Death Dis 2014; 5:e1210. [PMID: 24810046 PMCID: PMC4047872 DOI: 10.1038/cddis.2014.180] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2013] [Revised: 02/02/2014] [Accepted: 02/03/2014] [Indexed: 01/24/2023]
Abstract
There is currently no cure for advanced castration-refractory prostate cancer (CRPC) despite the recent approval of several new therapeutic agents. We report here the anti-tumor effect of the angio-inhibitory pigment epithelium-derived factor (PEDF) in the metastatic LNCaP-derivative CRPC CL1 model and explore PEDF anti-neoplasic efficacy in combination with low-dose chemotherapy. Androgen-sensitive LNCaP and CRPC PC3 cell lines were examined as comparison. Using a retroviral expression system, we showed that PEDF limited the proliferation of all prostatic cell lines tested; an effect attributed to interleukin 8 (IL8)-CXCR1/IL8RA inhibition. PEDF also reduced the number and size of 3D tumor spheroids in vitro, but only induced cell differentiation in CRPC spheroids. Similarly, PEDF inhibited the migration of CRPC cells suggesting both anti-proliferative and anti-migratory functions. In vivo, PEDF decreased by 85% and 65% the growth of subcutaneous (s.c.) PC3 and CL1 tumors, respectively. In the CL1 orthotopic model, tumor intake with lethal metastases was found in all animals; nevertheless, PEDF prolonged the median survival of tumor-bearing mice (95% confidence interval: 53±0.001 to 57±1 days). Accordingly, PEDF delayed the emergence of skeletal-related event in intra-tibial xenografts. Next, we evaluated low-dose docetaxel (DTX; 5, 1, 0.5 mg/kg) or cyclophosphamide (CTX; 10–20 mg/kg) on established s.c. PC3 tumors that conditionally express PEDF anti-tumoral epitope/NT3. Although NT3–DTX-5 mg/kg combination was inefficient, NT3–DTX-1 mg/kg and -0.5 mg/kg inhibited by 95% and 87.8%, respectively, tumor growth compared with control and induced tumor stasis. Both NT3–CTX combinations were advantageous. Inversely, PEDF–DTX-5 mg/kg and PEDF–CTX-10 mg/kg delayed the most CL1 tumor growth (15, 11 and 5 days for PEDF–DTX-5 mg/kg, PEDF–CTX-10 mg/kg and single treatments, respectively) with elevated apoptosis and serum thrombospondin-1 as possible mechanism and marker, respectively. As well, both PEDF–CTX-10 mg/kg and PEDF–DTX-5 mg/kg prolonged significantly the survival of tumor-bearing mice compared with single treatments. Metastases were reduced in PEDF–DTX-5 mg/kg compared with other treatments, suggesting that PEDF–DTX delayed metastases formation. Our results advocate that PEDF/low-dose chemotherapy may represent a new therapeutic alternative for CRPC.
Collapse
Affiliation(s)
- T Nelius
- Department of Urology, Texas Tech University-Health Sciences Center, 3601 4th Street, Lubbock, TX, USA
| | - D Martinez-Marin
- Department of Urology, Texas Tech University-Health Sciences Center, 3601 4th Street, Lubbock, TX, USA
| | - J Hirsch
- Department of Urology, Texas Tech University-Health Sciences Center, 3601 4th Street, Lubbock, TX, USA
| | - B Miller
- Department of Pathology, Texas Tech University-Health Sciences Center, 3601 4th Street, Lubbock, TX, USA
| | - K Rinard
- Department of Urology, Texas Tech University-Health Sciences Center, 3601 4th Street, Lubbock, TX, USA
| | - J Lopez
- Department of Urology, Texas Tech University-Health Sciences Center, 3601 4th Street, Lubbock, TX, USA
| | - W de Riese
- Department of Urology, Texas Tech University-Health Sciences Center, 3601 4th Street, Lubbock, TX, USA
| | - S Filleur
- 1] Department of Urology, Texas Tech University-Health Sciences Center, 3601 4th Street, Lubbock, TX, USA [2] Department of Immunology and Molecular Microbiology, Texas Tech University-Health Sciences Center, 3601 4th Street, Lubbock, TX, USA
| |
Collapse
|
27
|
Alcantara MB, Dass CR. Pigment epithelium-derived factor as a natural matrix metalloproteinase inhibitor: a comparison with classical matrix metalloproteinase inhibitors used for cancer treatment. J Pharm Pharmacol 2014; 66:895-902. [PMID: 24697787 DOI: 10.1111/jphp.12218] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 12/23/2013] [Indexed: 12/13/2022]
Abstract
OBJECTIVES In the 1990s, the discovery of the important role of matrix metalloproteinases (MMPs) in cancer angiogenesis, growth and metastasis galvanised research efforts to search for ways to inhibit these MMPs. To date, this has resulted in the investigation of approximately 50 MMPIs which have undergone various phases of clinical trials. However, despite a large body of research being devoted to discovery and development of MMPIs, results have largely not been supportive of this approach to anticancer treatment. KEY FINDINGS The reasons for the general failure of these drugs in clinical trials include various unwanted side-effects, the use of healthy volunteers to provide drug dosages which did not correctly reflect dosages for cancer patients, and the exclusion of patients with early stage cancer in clinical trials despite MMPs being determined to be critical for the angiogenic switch, a process associated with early tumour growth. In contrast, a naturally-occurring endogenous protein and a non-functional serine protease inhibitor (serpin), pigment epithelium-derived factor (PEDF), has been proposed for cancer therapy partly due to its ability to regulate specific MMPs central to cancer progression. SUMMARY PEDF has been found to specifically downregulate membrane-type I matrix metalloproteinase (MT1-MMP) and furthermore, potentially matrix metalloproteinase-2 (MMP-2), two of the most commonly implicated MMPs in neoplasia.
Collapse
Affiliation(s)
- Marice B Alcantara
- College of Health and Biomedicine, Victoria University, St Albans, Australia
| | | |
Collapse
|
28
|
Overcoming intratumor heterogeneity of polygenic cancer drug resistance with improved biomarker integration. Neoplasia 2013; 14:1278-89. [PMID: 23308059 DOI: 10.1593/neo.122096] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 12/11/2012] [Accepted: 12/11/2012] [Indexed: 12/14/2022] Open
Abstract
Improvements in technology and resources are helping to advance our understanding of cancer-initiating events as well as factors involved with tumor progression, adaptation, and evasion of therapy. Tumors are well known to contain diverse cell populations and intratumor heterogeneity affords neoplasms with a diverse set of biologic characteristics that can be used to evolve and adapt. Intratumor heterogeneity has emerged as a major hindrance to improving cancer patient care. Polygenic cancer drug resistance necessitates reconsidering drug designs to include polypharmacology in pursuit of novel combinatorial agents having multitarget activity to overcome the diverse and compensatory signaling pathways in which cancer cells use to survive and evade therapy. Advances will require integration of different biomarkers such as genomics and imaging to provide for more adequate elucidation of the spatially varying location, type, and extent of diverse intratumor signaling molecules to provide for a rationale-based personalized cancer medicine strategy.
Collapse
|
29
|
Becerra SP, Notario V. The effects of PEDF on cancer biology: mechanisms of action and therapeutic potential. Nat Rev Cancer 2013; 13:258-71. [PMID: 23486238 PMCID: PMC3707632 DOI: 10.1038/nrc3484] [Citation(s) in RCA: 162] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The potent actions of pigment epithelium-derived factor (PEDF) on tumour-associated cells, and its extracellular localization and secretion, stimulated research on this multifunctional serpin. Such studies have identified several PEDF receptors and downstream signalling pathways. Known cellular PEDF responses have expanded from the initial discovery that PEDF induces retinoblastoma cell differentiation to its anti-angiogenic, antitumorigenic and antimetastatic properties. Although the diversity of PEDF activities seems to be complex, they are consistent with the varied mechanisms that regulate this multimodal factor. If PEDF is to be used for cancer management, a deeper appreciation of its many functions and mechanisms of action is needed.
Collapse
Affiliation(s)
- S Patricia Becerra
- National Eye Institute, US National Institutes of Health, Bethesda, Maryland, USA.
| | | |
Collapse
|
30
|
Nelius T, Samathanam C, Martinez-Marin D, Gaines N, Stevens J, Hickson J, de Riese W, Filleur S. Positive correlation between PEDF expression levels and macrophage density in the human prostate. Prostate 2013; 73:549-61. [PMID: 23038613 PMCID: PMC3600115 DOI: 10.1002/pros.22595] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Accepted: 09/04/2012] [Indexed: 02/03/2023]
Abstract
BACKGROUND In this study, we investigated the capacity of pigment epithelium-derived factor (PEDF) to modulate the recruitment and the differentiation of monocytes/macrophages both in vitro and in human prostate. METHODS Using Boyden chambers, we assessed PEDF effect on the migration of monocytes and chemically activated RAW 264.7 macrophages. Normal, prostatitis, and prostate cancer specimens were retrospectively selected and examined by immunohistochemistry for PEDF expression and infiltration of immune CD68 + macrophagic cells. PEDF expression and macrophage density were then correlated with each other and clinicopathological parameters. M1 and M2 differentiation markers were quantified by qRT-PCR, Western blotting, and ELISA. RESULTS In chemotaxis, PEDF induced the migration of monocytes/macrophages. In immunohistochemistry, macrophages were markedly increased in prostatitis and malignant compared to normal tissues. PEDF was expressed at variable levels in the stroma and epithelium. PEDF mRNA was down-regulated in both prostate cancer and prostatitis compared to normal tissues. In correlation studies, macrophage density and PEDF expression were respectively positively and negatively associated with prostate size. Most importantly, PEDF expression positively correlated with macrophage density. Finally, PEDF stimulated the expression of iNOS, IL12, and TNFα; and inhibited IL10 and arginase 1 in mouse and human macrophages confirming a M1-type differentiation. CONCLUSIONS Our data demonstrate that PEDF acts directly on monocytes/macrophages by inducing their migration and differentiation into M1-type cells. These findings suggest a possible role of macrophages in PEDF anti-tumor properties and may support further development of PEDF-based anti-cancer therapy.
Collapse
Affiliation(s)
- Thomas Nelius
- Texas Tech University Health Sciences Center, Department of Urology, Lubbock-TX
| | - Christina Samathanam
- The University of Texas Health Science Center at Houston, Medical School, Department of Pathology and Laboratory Medicine, Houston-TX
| | | | - Natalie Gaines
- Texas Tech University Health Sciences Center, Department of Urology, Lubbock-TX
| | - Jessica Stevens
- Texas Tech University Health Sciences Center, Department of Urology, Lubbock-TX
| | - Johnny Hickson
- Texas Tech University Health Sciences Center, Department of Urology, Lubbock-TX
| | - Werner de Riese
- Texas Tech University Health Sciences Center, Department of Urology, Lubbock-TX
| | - Stéphanie Filleur
- Texas Tech University Health Sciences Center, Department of Urology, Lubbock-TX
- Texas Tech University Health Sciences Center, Department of Immunology and Molecular Microbiology, Lubbock-TX
| |
Collapse
|
31
|
Jia L, Waxman DJ. Thrombospondin-1 and pigment epithelium-derived factor enhance responsiveness of KM12 colon tumor to metronomic cyclophosphamide but have disparate effects on tumor metastasis. Cancer Lett 2012; 330:241-9. [PMID: 23228633 DOI: 10.1016/j.canlet.2012.11.055] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 11/30/2012] [Accepted: 11/30/2012] [Indexed: 12/18/2022]
Abstract
The anti-tumor activity, metronomic chemotherapy sensitization potential and metastatic effects of the endogenous angiogenesis inhibitors thrombospondin-1 and PEDF were investigated in KM12 colon adenocarcinoma xenografts. Thrombospondin-1 and PEDF decreased KM12 tumor microvessel density, increased macrophage infiltration, and improved responsiveness to metronomic cyclophosphamide (CPA) treatment, but did not activate the anti-tumor innate immunity that metronomic CPA induces in other tumor models. Moreover, thrombospondin-1, but not PEDF, significantly increased KM12 metastasis to the lung, while PEDF augmented the anti-metastatic activity of metronomic CPA. Thus, while thrombospondin-1 and PEDF both increase the KM12 tumor responsiveness to metronomic CPA, they have disparate effects on tumor metastasis.
Collapse
Affiliation(s)
- Li Jia
- Division of Cell and Molecular Biology, Department of Biology, Boston University, Boston, MA 02215, United States
| | | |
Collapse
|
32
|
Li L, Yang J, Wang WW, Yao YC, Fang SH, Dai ZY, Hong HH, Yang X, Shuai XT, Gao GQ. Pigment epithelium-derived factor gene loaded in cRGD–PEG–PEI suppresses colorectal cancer growth by targeting endothelial cells. Int J Pharm 2012; 438:1-10. [DOI: 10.1016/j.ijpharm.2012.08.043] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Revised: 07/30/2012] [Accepted: 08/21/2012] [Indexed: 01/19/2023]
|
33
|
The interconnectedness of cancer cell signaling. Neoplasia 2012; 13:1183-93. [PMID: 22241964 DOI: 10.1593/neo.111746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Revised: 12/14/2011] [Accepted: 12/14/2011] [Indexed: 11/18/2022] Open
Abstract
The elegance of fundamental and applied research activities have begun to reveal a myriad of spatial and temporal alterations in downstream signaling networks affected by cell surface receptor stimulation including G protein-coupled receptors and receptor tyrosine kinases. Interconnected biochemical pathways serve to integrate and distribute the signaling information throughout the cell by orchestration of complex biochemical circuits consisting of protein interactions and covalent modification processes. It is clear that scientific literature summarizing results from both fundamental and applied scientific research activities has served to provide a broad foundational biologic database that has been instrumental in advancing our continued understanding of underlying cancer biology. This article reflects on historical advances and the role of innovation in the competitive world of grant-sponsored research.
Collapse
|
34
|
Doloff JC, Waxman DJ. VEGF receptor inhibitors block the ability of metronomically dosed cyclophosphamide to activate innate immunity-induced tumor regression. Cancer Res 2012; 72:1103-15. [PMID: 22237627 DOI: 10.1158/0008-5472.can-11-3380] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
In metronomic chemotherapy, frequent drug administration at lower than maximally tolerated doses can improve activity while reducing the dose-limiting toxicity of conventional dosing schedules. Although the antitumor activity produced by metronomic chemotherapy is attributed widely to antiangiogenesis, the significance of this mechanism remains somewhat unclear. In this study, we show that a 6-day repeating metronomic schedule of cyclophosphamide administration activates a potent antitumor immune response associated with brain tumor recruitment of natural killer (NK) cells, macrophages, and dendritic cells that leads to marked tumor regression. Tumor regression was blocked in nonobese diabetic/severe combined immunodeficient (NOD/SCID-γ) mice, which are deficient or dysfunctional in all these immune cell types. Furthermore, regression was blunted by NK cell depletion in immunocompetent syngeneic mice or in perforin-deficient mice, which are compromised for NK, NKT, and T-cell cytolytic functions. Unexpectedly, we found that VEGF receptor inhibitors blocked both innate immune cell recruitment and the associated tumor regression response. Cyclophosphamide administered at a maximum tolerated dose activated a transient, weak innate immune response, arguing that persistent drug-induced cytotoxic damage or associated cytokine and chemokine responses are required for effective innate immunity-based tumor regression. Together, our results reveal an innate immunity-based mechanism of tumor regression that can be activated by a traditional cytotoxic chemotherapy administered on a metronomic schedule. These findings suggest the need to carefully evaluate the clinical effects of combination chemotherapies that incorporate antiangiogenesis drugs targeting VEGF receptor.
Collapse
Affiliation(s)
- Joshua C Doloff
- Division of Cell and Molecular Biology, Department of Biology, Boston University, Boston, Massachusetts 02215, USA
| | | |
Collapse
|
35
|
Josefsson A, Adamo H, Hammarsten P, Granfors T, Stattin P, Egevad L, Laurent AE, Wikström P, Bergh A. Prostate cancer increases hyaluronan in surrounding nonmalignant stroma, and this response is associated with tumor growth and an unfavorable outcome. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:1961-8. [PMID: 21854754 DOI: 10.1016/j.ajpath.2011.06.005] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2010] [Revised: 05/02/2011] [Accepted: 06/28/2011] [Indexed: 01/01/2023]
Abstract
Our objective was to investigate whether the presence of a tumor increases hyaluronan (HA) levels in surrounding prostate tissues and whether this extratumoral HA influences tumor growth and outcome. From a series of 287 men diagnosed with prostate cancer at transurethral resection and followed up with watchful waiting, tissue microarrays were constructed, stained, and scored for HA. A high HA staining score in the tumor stroma or in nonmalignant prostate tissue stroma were both associated positively with higher Gleason score and larger tumor volume, and was associated with a poor outcome. HA staining score was not an independent marker for outcome (multivariate Cox, with Gleason score, tumor volume, stage, and HA variables). In an orthotopic rat prostate cancer model, hyaluronic acid synthase-1 mRNA levels and HA staining were increased in normal prostate tissue surrounding prostate cancer. Orthotopic prostate cancer growth was increased by intraprostatic injection of HA. In conclusion, cancer in the prostate apparently stimulates HA synthesis both in tumor stroma and in the surrounding normal tissue. This promoted tumor growth and was associated with an unfavorable outcome.
Collapse
Affiliation(s)
- Andreas Josefsson
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden.
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Ho TC, Chen SL, Shih SC, Chang SJ, Yang SL, Hsieh JW, Cheng HC, Chen LJ, Tsao YP. Pigment epithelium-derived factor (PEDF) promotes tumor cell death by inducing macrophage membrane tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). J Biol Chem 2011; 286:35943-35954. [PMID: 21846721 DOI: 10.1074/jbc.m111.266064] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pigment epithelium-derived factor (PEDF) is an intrinsic anti-angiogenic factor and a potential anti-tumor agent. The tumoricidal mechanism of PEDF, however, has not been fully elucidated. Here we report that PEDF induces the apoptosis of TC-1 and SK-Hep-1 tumor cells when they are cocultured with bone marrow-derived macrophages (BMDMs). This macrophage-mediated tumor killing is prevented by blockage of TNF-related apoptosis-inducing ligand (TRAIL) following treatment with the soluble TRAIL receptor. PEDF also increases the amount of membrane-bound TRAIL on cultured mouse BMDMs and on macrophages surrounding subcutaneous tumors. PEDF-induced tumor killing and TRAIL induction are abrogated by peroxisome proliferator-activated receptor γ (PPARγ) antagonists or small interfering RNAs targeting PPARγ. PEDF also induces PPARγ in BMDMs. Furthermore, the activity of the TRAIL promoter in human macrophages is increased by PEDF stimulation. Chromatin immunoprecipitation and DNA pull-down assays confirmed that endogenous PPARγ binds to a functional PPAR-response element (PPRE) in the TRAIL promoter, and mutation of this PPRE abolishes the binding of the PPARγ-RXRα heterodimer. Also, PPARγ-dependent transactivation and PPARγ-RXRα binding to this PPRE are prevented by PPARγ antagonists. Our results provide a novel mechanism for the tumoricidal activity of PEDF, which involves tumor cell killing via PPARγ-mediated TRAIL induction in macrophages.
Collapse
Affiliation(s)
- Tsung-Chuan Ho
- Department of Medical Research, Mackay Memorial Hospital, Taipei 104, Taiwan
| | - Show-Li Chen
- Department of Microbiology, School of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Shou-Chuan Shih
- Department of Gastroenterology, Mackay Memorial Hospital, Taipei 104, Taiwan; Mackay Medicine, Nursing, and Management College, Taipei 112, Taiwan
| | - Shing-Jyh Chang
- Department of Obstetrics and Gynecology, Mackay Memorial Hospital, Taipei 104, Taiwan; Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Su-Lin Yang
- Centers for Disease Control, Taipei 115, Taiwan; Department of Microbiology and Immunology, National Defense Medical Center, Taipei 114, Taiwan
| | - Jui-Wen Hsieh
- Mackay Medicine, Nursing, and Management College, Taipei 112, Taiwan; Department of Ophthalmology, Mackay Memorial Hospital, Taipei 104, Taiwan
| | - Huey-Chuan Cheng
- Department of Ophthalmology, Mackay Memorial Hospital, Taipei 104, Taiwan
| | - Lee-Jen Chen
- Mackay Medicine, Nursing, and Management College, Taipei 112, Taiwan; Department of Ophthalmology, Mackay Memorial Hospital, Taipei 104, Taiwan
| | - Yeou-Ping Tsao
- Department of Medical Research, Mackay Memorial Hospital, Taipei 104, Taiwan; Department of Microbiology and Immunology, National Defense Medical Center, Taipei 114, Taiwan; Department of Ophthalmology, Mackay Memorial Hospital, Taipei 104, Taiwan.
| |
Collapse
|
37
|
Dinosaurs and ancient civilizations: reflections on the treatment of cancer. Neoplasia 2011; 12:957-68. [PMID: 21170260 DOI: 10.1593/neo.101588] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2010] [Revised: 11/15/2010] [Accepted: 11/15/2010] [Indexed: 12/14/2022] Open
Abstract
Research efforts in the area of palaeopathology have been seen as an avenue to improve our understanding of the pathogenesis of cancer. Answers to questions of whether dinosaurs had cancer, or if cancer plagued ancient civilizations, have captured the imagination as well as the popular media. Evidence for dinosaurian cancer may indicate that cancer may have been with us from the dawn of time. Ancient recorded history suggests that past civilizations attempted to fight cancer with a variety of interventions. When contemplating the issue why a generalized cure for cancer has not been found, it might prove useful to reflect on the relatively limited time that this issue has been an agenda item of governmental attention as well as continued introduction of an every evolving myriad of manmade carcinogens relative to the total time cancer has been present on planet Earth. This article reflects on the history of cancer and the progress made following the initiation of the "era of cancer chemotherapy."
Collapse
|
38
|
Halin S, Hammarsten P, Adamo H, Wikström P, Bergh A. Tumor indicating normal tissue could be a new source of diagnostic and prognostic markers for prostate cancer. ACTA ACUST UNITED AC 2010; 5:37-47. [PMID: 23484475 DOI: 10.1517/17530059.2011.540009] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
IMPORTANCE OF THE FIELD Prostate cancer is a common and multifocal disease but the diagnostic methods available are unsatisfactory. Most tumors present are of low malignant potential, whereas others are highly aggressive. At present, imaging cannot be used to guide tissue biopsies safely towards the most aggressive tumor present. To handle this problem multiple needle biopsies are taken. The biopsies often contain only normal prostate tissue, and even if the tumor is sampled it is not known whether a more aggressive cancer is present elsewhere in the organ. If changes in the normal tissue indicate the presence and nature of tumors, this information could be used to improve diagnostics and prognostics of prostate cancer. AREAS COVERED IN THIS REVIEW Current evidence that the tumor-adjacent morphologically normal prostate tissue is not completely normal is reviewed, and that this tissue, named tumor indicating normal tissue (TINT) by the authors, can be used to diagnose prostate cancer. WHAT THE READER WILL GAIN The reader will understand that tumors need to affect their surroundings in order to grow and metastasize and that the normal prostate tissue is therefore tinted by the presence and nature of cancer and that this knowledge can be used to develop new diagnostic and prognostic markers. TAKE HOME MESSAGE TINT changes could probably, when more rigorously defined and validated, be used to diagnose and prognosticate prostate cancer.
Collapse
Affiliation(s)
- Sofia Halin
- Umeå University, Department of Medical Biosciences, Pathology, Building 6M, Second Floor, S-90187 Umeå, Sweden +46 90 785 15 30 ; +46 90 785 44 84 ;
| | | | | | | | | |
Collapse
|