1
|
Ghosh S, Mahajan AA, Dey A, Rajendran RL, Chowdhury A, Sen S, Paul S, Majhi S, Hong CM, Gangadaran P, Ahn BC, Krishnan A. Exosomes in Bone Cancer: Unveiling their Vital Role in Diagnosis, Prognosis, and Therapeutic Advancements. J Cancer 2024; 15:4128-4142. [PMID: 38947401 PMCID: PMC11212077 DOI: 10.7150/jca.95709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 05/19/2024] [Indexed: 07/02/2024] Open
Abstract
Bone cancer among adolescents and children exhibits varying survival outcomes based on disease state. While localized bone cancer cases have a survival rate exceeding 70%, metastatic, refractory, and recurrent forms are associated with significantly poorer prognoses. Initially believed to be mere vehicles for cellular waste disposal, exosomes are now recognized as extracellular vesicles facilitating intercellular communication. These vesicles influence cellular behaviors by transporting various biomolecules, such as proteins, DNA, RNA, and lipids, among cells. The role of exosomes in regulating the progression of bone cancer is increasingly evident, impacting critical processes like tumorigenesis, proliferation, metastasis, angiogenesis, immune evasion, and drug resistance. Current research underscores the substantial potential of exosomes in promoting the progression and development of bone cancer. This review delves into the complex process of exosome biogenesis, the variety of cell-derived exosome sources, and their applications in drug delivery and therapeutics. It also examines ongoing clinical trials focused on exosome cargo levels and discusses the challenges and future directions in exosome research. Unlike costly and invasive traditional diagnostic methods, exosomal biomarkers offer a non-invasive, cost-effective, and readily accessible routine screening through simple fluid collection that aims to inspire researchers to investigate the potential of exosomes for cancer theragnostic. Through comprehensive exploration of these areas, the review seeks to enhance understanding and foster innovative solutions to cancer biology in the near future.
Collapse
Affiliation(s)
- Subhrojyoti Ghosh
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| | - Atharva Anand Mahajan
- Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Mumbai, Maharashtra 410210, India
| | - Anuvab Dey
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, North Guwahati, Assam 781039, India
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Ankita Chowdhury
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Delhi 110016, India
| | - Sushmita Sen
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Delhi 110016, India
| | - Subhobrata Paul
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Delhi 110016, India
| | - Sourav Majhi
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Delhi 110016, India
| | - Chae Moon Hong
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu 41944, Republic of Korea
| | - Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu 41944, Republic of Korea
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Anand Krishnan
- Department of Chemical Pathology, School of Pathology, Faculty of Health Sciences, University of the Free State, Bloemfontein, 9300, South Africa
| |
Collapse
|
2
|
Peyronnel C, Totoson P, Martin H, Demougeot C. Relevance of circulating markers of endothelial activation for cardiovascular risk assessment in rheumatoid arthritis: a narrative review. Life Sci 2023; 314:121264. [PMID: 36470540 DOI: 10.1016/j.lfs.2022.121264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 11/22/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
Rheumatoid arthritis (RA) is associated with excessive cardiovascular mortality secondary to premature atherosclerosis, in which endothelial activation (EA) plays a central role. EA is characterized by loss of vascular integrity, expression of leucocyte adhesion molecules, transition from antithrombotic to prothrombotic phenotype, cytokines production, shedding of membrane microparticles and recruitment of endothelial progenitor cells. As EA is an early event in atherogenesis, circulating markers of EA are putative markers of vascular pathology and cardiovascular (CV) risk. After a presentation of biology of EA, the present review analyzed the available data regarding changes in EA markers in RA in link with the vascular pathology and CV events, discussed their relevance as biomarkers of CV risk and proposed future directions.
Collapse
Affiliation(s)
- Célian Peyronnel
- PEPITE EA 4267, Université de Franche-Comté, F-25000 Besançon, France
| | - Perle Totoson
- PEPITE EA 4267, Université de Franche-Comté, F-25000 Besançon, France
| | - Hélène Martin
- PEPITE EA 4267, Université de Franche-Comté, F-25000 Besançon, France
| | - Céline Demougeot
- PEPITE EA 4267, Université de Franche-Comté, F-25000 Besançon, France.
| |
Collapse
|
3
|
Kumar A, Sood A, Singhmar R, Mishra YK, Thakur VK, Han SS. Manufacturing functional hydrogels for inducing angiogenic-osteogenic coupled progressions in hard tissue repairs: prospects and challenges. Biomater Sci 2022; 10:5472-5497. [PMID: 35994005 DOI: 10.1039/d2bm00894g] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
In large bone defects, inadequate vascularization within the engineered constructs has been a major challenge in developing clinically impactful products. It is fairly determined that bone tissues and blood vessels are established concurrently throughout tissue repairs after an injury. Thus, the coupling of angiogenesis-osteogenesis is an essential course of action in bone tissue restoration. The manufacture of biomaterial-based scaffolds plays a decisive role in stimulating angiogenic and osteogenic progressions (instruction of neovascularization and bone mineralization). Bone hydrogels with optimal conditions are more efficient at healing bone defects. There has been a remarkable advancement in producing bone substitutes in the tissue engineering area, but the sufficient and timely vascularization of engineered constructs for optimal tissue integration and regeneration is lacking due to mismatch in the scaffold characteristics and new bone tissue reconstruction. Therefore, various key challenges remain to be overcome. A deep understanding of angiogenesis and osteogenesis progressions is required to manufacture bone hydrogels with satisfactory results. The current review briefly discusses the fundamentals of bone tissues, the significance of angiogenesis-osteogenesis progressions and their inducers, the efficacy of biomaterials and composite hydrogel-promoted neo-vasculogenesis (i.e. angiogenesis) and bone mineralization (i.e. osteogenesis), and related challenges, including future research directions.
Collapse
Affiliation(s)
- Anuj Kumar
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea. .,Research Institute of Cell Culture, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea
| | - Ankur Sood
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea.
| | - Ritu Singhmar
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea.
| | - Yogendra Kumar Mishra
- Smart Materials, NanoSYD, Mads Clausen Institute, University of Southern Denmark, Alsion 2, 6400, Sønderborg, Denmark
| | - Vijay Kumar Thakur
- Biorefining and Advanced Materials Research Center, Scotland's Rural College (SRUC), Kings Buildings, Edinburgh EH9 3JG, UK.,School of Engineering, University of Petroleum and Energy Studies (UPES), Dehradun 248007, Uttarakhand, India
| | - Sung Soo Han
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea. .,Research Institute of Cell Culture, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, South Korea
| |
Collapse
|
4
|
Rubin JB, Lagas JS, Broestl L, Sponagel J, Rockwell N, Rhee G, Rosen SF, Chen S, Klein RS, Imoukhuede P, Luo J. Sex differences in cancer mechanisms. Biol Sex Differ 2020; 11:17. [PMID: 32295632 PMCID: PMC7161126 DOI: 10.1186/s13293-020-00291-x] [Citation(s) in RCA: 149] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 03/18/2020] [Indexed: 02/07/2023] Open
Abstract
We now know that cancer is many different diseases, with great variation even within a single histological subtype. With the current emphasis on developing personalized approaches to cancer treatment, it is astonishing that we have not yet systematically incorporated the biology of sex differences into our paradigms for laboratory and clinical cancer research. While some sex differences in cancer arise through the actions of circulating sex hormones, other sex differences are independent of estrogen, testosterone, or progesterone levels. Instead, these differences are the result of sexual differentiation, a process that involves genetic and epigenetic mechanisms, in addition to acute sex hormone actions. Sexual differentiation begins with fertilization and continues beyond menopause. It affects virtually every body system, resulting in marked sex differences in such areas as growth, lifespan, metabolism, and immunity, all of which can impact on cancer progression, treatment response, and survival. These organismal level differences have correlates at the cellular level, and thus, males and females can fundamentally differ in their protections and vulnerabilities to cancer, from cellular transformation through all stages of progression, spread, and response to treatment. Our goal in this review is to cover some of the robust sex differences that exist in core cancer pathways and to make the case for inclusion of sex as a biological variable in all laboratory and clinical cancer research. We finish with a discussion of lab- and clinic-based experimental design that should be used when testing whether sex matters and the appropriate statistical models to apply in data analysis for rigorous evaluations of potential sex effects. It is our goal to facilitate the evaluation of sex differences in cancer in order to improve outcomes for all patients.
Collapse
Affiliation(s)
- Joshua B Rubin
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA.
- Department of Neuroscience, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA.
| | - Joseph S Lagas
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Lauren Broestl
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Jasmin Sponagel
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Nathan Rockwell
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Gina Rhee
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Sarah F Rosen
- Department of Medicine, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Si Chen
- Department of Biomedical Engineering, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Robyn S Klein
- Department of Neuroscience, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
- Department of Medicine, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Princess Imoukhuede
- Department of Biomedical Engineering, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Jingqin Luo
- Department of Surgery, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| |
Collapse
|
5
|
Schreier S, Triampo W. The Blood Circulating Rare Cell Population. What is it and What is it Good For? Cells 2020; 9:cells9040790. [PMID: 32218149 PMCID: PMC7226460 DOI: 10.3390/cells9040790] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/13/2020] [Accepted: 03/16/2020] [Indexed: 02/06/2023] Open
Abstract
Blood contains a diverse cell population of low concentration hematopoietic as well as non-hematopoietic cells. The majority of such rare cells may be bone marrow-derived progenitor and stem cells. This paucity of circulating rare cells, in particular in the peripheral circulation, has led many to believe that bone marrow as well as other organ-related cell egress into the circulation is a response to pathological conditions. Little is known about this, though an increasing body of literature can be found suggesting commonness of certain rare cell types in the peripheral blood under physiological conditions. Thus, the isolation and detection of circulating rare cells appears to be merely a technological problem. Knowledge about rare cell types that may circulate the blood stream will help to advance the field of cell-based liquid biopsy by supporting inter-platform comparability, making use of biological correct cutoffs and “mining” new biomarkers and combinations thereof in clinical diagnosis and therapy. Therefore, this review intends to lay ground for a comprehensive analysis of the peripheral blood rare cell population given the necessity to target a broader range of cell types for improved biomarker performance in cell-based liquid biopsy.
Collapse
Affiliation(s)
- Stefan Schreier
- School of Bioinnovation and Bio-based Product Intelligence, Faculty of Science, Mahidol University, Rama VI Rd, Bangkok 10400, Thailand;
- Thailand Center of Excellence in Physics, Ministry of Higher Education, Science, Research and Innovation, 328 Si Ayutthaya Road, Bangkok 10400, Thailand
| | - Wannapong Triampo
- Thailand Center of Excellence in Physics, Ministry of Higher Education, Science, Research and Innovation, 328 Si Ayutthaya Road, Bangkok 10400, Thailand
- Department of Physics, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
- Correspondence:
| |
Collapse
|
6
|
Hewes JL, Lee JY, Fagan KA, Bauer NN. The changing face of pulmonary hypertension diagnosis: a historical perspective on the influence of diagnostics and biomarkers. Pulm Circ 2020; 10:2045894019892801. [PMID: 32110383 PMCID: PMC7000867 DOI: 10.1177/2045894019892801] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 11/11/2019] [Indexed: 12/15/2022] Open
Abstract
Pulmonary hypertension is a complex, multifactorial disease that results in right heart failure and premature death. Since the initial reports of pulmonary hypertension in the late 1800s, the diagnosis of pulmonary hypertension has evolved with respect to its definition, screening tools, and diagnostic techniques. This historical perspective traces the earliest roots of pulmonary hypertension detection and diagnosis through to the current recommendations for classification. We highlight the diagnostic tools used in the past and present, and end with a focus on the future directions of early detection. Early detection of pulmonary hypertension and pulmonary arterial hypertension and the proper determination of etiology are vital for the early therapeutic intervention that can prolong life expectancy and improve quality of life. The search for a non-invasive screening tool for the identification and classification of pulmonary hypertension is ongoing, and we discuss the role of animal models of the disease in this search.
Collapse
Affiliation(s)
- Jenny L. Hewes
- Department of Pharmacology, College of
Medicine,
University
of South Alabama, Mobile, AL, USA
- Center for Lung Biology, College of
Medicine,
University
of South Alabama, Mobile, AL, USA
| | - Ji Young Lee
- Center for Lung Biology, College of
Medicine,
University
of South Alabama, Mobile, AL, USA
- Division of Pulmonary and Critical Care
Medicine, University Hospital,
University
of South Alabama, Mobile, AL, USA
- Department of Physiology and Cell
Biology, College of Medicine,
University
of South Alabama, Mobile, AL, USA
| | - Karen A. Fagan
- Department of Pharmacology, College of
Medicine,
University
of South Alabama, Mobile, AL, USA
- Center for Lung Biology, College of
Medicine,
University
of South Alabama, Mobile, AL, USA
- Division of Pulmonary and Critical Care
Medicine, University Hospital,
University
of South Alabama, Mobile, AL, USA
| | - Natalie N. Bauer
- Department of Pharmacology, College of
Medicine,
University
of South Alabama, Mobile, AL, USA
- Center for Lung Biology, College of
Medicine,
University
of South Alabama, Mobile, AL, USA
| |
Collapse
|
7
|
Capozzi M, De Divitiis C, Ottaiano A, von Arx C, Scala S, Tatangelo F, Delrio P, Tafuto S. Lenvatinib, a molecule with versatile application: from preclinical evidence to future development in anti-cancer treatment. Cancer Manag Res 2019; 11:3847-3860. [PMID: 31118801 PMCID: PMC6502442 DOI: 10.2147/cmar.s188316] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 02/28/2019] [Indexed: 12/13/2022] Open
Abstract
Lenvatinib is an emerging multi-kinase inhibitor with a preferential anti-angiogenic activity, which has shown efficacy in the treatment of renal cell carcinoma, differentiated thyroid cancer and hepatocellular carcinoma. It inhibits vascular endothelial growth factor receptor family (VEGFR1–3), fibroblast growth factor receptor family (FGFR1–4), platelet-derived growth factor receptor–alpha (PDGFRα), tyrosine-kinase receptor (KIT) and rearranged during transfection receptor (RET). In this review we have evaluated the development from bench to bedside of lenvatinib. PubMed, MEDLINE and clinicaltrials.gov are the sources of data. Furthermore, the preclinical in vitro and in vivo data, as well as efficacy and toxicity results of lenvatinib in the clinic, are presented and discussed. Treatment with lenvatinib causes side effects (hypertension, proteinuria, fatigue and diarrhea), which are predominantly related to the inhibition of angiogenesis. For these reasons, the identification of biomarkers of efficacy and resistance to lenvatinib is a key challenge in order to select responsive patients. This review provides an overview on lenvatinib's clinical use, perspectives and indications for future development.
Collapse
Affiliation(s)
- Monica Capozzi
- Department of Abdominal Oncology, Istituto Nazionale Tumori, IRCCS - Fondazione "G. Pascale", Napoli, Italia
| | - Chiara De Divitiis
- UOSD Oncology- AOU "San Giovanni di Dio e Ruggi D'Aragona", Salerno, Italia
| | - Alessandro Ottaiano
- SSD Innovative Therapies for Abdominal Metastases - Department of Abdominal Oncology, Istituto Nazionale Tumori, IRCCS - Fondazione "G. Pascale", Napoli, Italia
| | - Claudia von Arx
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Stefania Scala
- Molecular Immunology and Immunoregulation, Istituto Nazionale Tumori, IRCCS - Fondazione "G. Pascale", Napoli, Italia
| | - Fabiana Tatangelo
- Department of Pathology, Istituto Nazionale Tumori, IRCCS - Fondazione "G. Pascale", Napoli, Italia
| | - Paolo Delrio
- Department of Abdominal Oncology, Istituto Nazionale Tumori, IRCCS - Fondazione "G. Pascale", Napoli, Italia
| | - Salvatore Tafuto
- Department of Abdominal Oncology, Istituto Nazionale Tumori, IRCCS - Fondazione "G. Pascale", Napoli, Italia
| |
Collapse
|
8
|
McMahon D, Mah E, Hynynen K. Angiogenic response of rat hippocampal vasculature to focused ultrasound-mediated increases in blood-brain barrier permeability. Sci Rep 2018; 8:12178. [PMID: 30111814 PMCID: PMC6093874 DOI: 10.1038/s41598-018-30825-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 08/06/2018] [Indexed: 01/12/2023] Open
Abstract
Focused ultrasound (FUS) and circulating microbubbles can induce a targeted and transient increase in blood-brain barrier permeability. While preclinical research has demonstrated the utility of FUS for efficacious drug deliver to the brain, there remain gaps in our knowledge regarding the long-term response of brain vasculature to this intervention. Previous work has demonstrated transcriptional changes in hippocampal microvessels following sonication that are indicative of the initiation of angiogenic processes. Moreover, blood vessel growth has been reported in skeletal muscle following application of FUS and microbubbles. The current study demonstrates that blood vessel density in the rat hippocampus is modestly elevated at 7 and 14 d post-FUS compared to the contralateral hemisphere (7 d: 10.9 ± 6.0%, p = 0.02; 14 d: 12.1 ± 3.2%, p < 0.01), but returns to baseline by 21 d (5.9 ± 2.6%, p = 0.12). Concurrently, relative newborn endothelial cell density and frequency of small blood vessel segments were both elevated in the sonicated hippocampus. While further work is required to determine the mechanisms driving these changes, the findings presented here may have relevance to the optimal frequency of repeated treatments.
Collapse
Affiliation(s)
- Dallan McMahon
- Sunnybrook Research Institute, Toronto, M4N 3M5, Canada. .,University of Toronto, Department of Medical Biophysics, Toronto, M4N 3M5, Canada.
| | - Ethan Mah
- Sunnybrook Research Institute, Toronto, M4N 3M5, Canada
| | - Kullervo Hynynen
- Sunnybrook Research Institute, Toronto, M4N 3M5, Canada.,University of Toronto, Department of Medical Biophysics, Toronto, M4N 3M5, Canada.,University of Toronto, Institute of Biomaterials and Biomedical Engineering, Toronto, M5S 3G9, Canada
| |
Collapse
|
9
|
Dykstra JA, Facile T, Patrick RJ, Francis KR, Milanovich S, Weimer JM, Kota DJ. Concise Review: Fat and Furious: Harnessing the Full Potential of Adipose-Derived Stromal Vascular Fraction. Stem Cells Transl Med 2017; 6:1096-1108. [PMID: 28186685 PMCID: PMC5388064 DOI: 10.1002/sctm.16-0337] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 10/14/2016] [Accepted: 11/07/2016] [Indexed: 12/28/2022] Open
Abstract
Due to their capacity to self-renew, proliferate and generate multi-lineage cells, adult-derived stem cells offer great potential for use in regenerative therapies to stop and/or reverse degenerative diseases such as diabetes, heart failure, Alzheimer's disease and others. However, these subsets of cells can be isolated from different niches, each with differing potential for therapeutic applications. The stromal vascular fraction (SVF), a stem cell enriched and adipose-derived cell population, has garnered interest as a therapeutic in regenerative medicine due to its ability to secrete paracrine factors that accelerate endogenous repair, ease of accessibility and lack of identified major adverse effects. Thus, one can easily understand the rush to employ adipose-derived SVF to treat human disease. Perhaps faster than any other cell preparation, SVF is making its way to clinics worldwide, while critical preclinical research needed to establish SVF safety, efficacy and optimal, standardized clinical procedures are underway. Here, we will provide an overview of the current knowledge driving this phenomenon, its regulatory issues and existing studies, and propose potential unmapped applications. Stem Cells Translational Medicine 2017;6:1096-1108.
Collapse
Affiliation(s)
- Jordan A. Dykstra
- Children's Health Research Center, Sanford Research, Sioux FallsSouth DakotaUSA
| | - Tiffany Facile
- Children's Health Research Center, Sanford Research, Sioux FallsSouth DakotaUSA
| | - Ryan J. Patrick
- Children's Health Research Center, Sanford Research, Sioux FallsSouth DakotaUSA
| | - Kevin R. Francis
- Children's Health Research Center, Sanford Research, Sioux FallsSouth DakotaUSA
- Department of PediatricsThe University of South Dakota Sanford School of MedicineVermillion, South DakotaUSA
| | - Samuel Milanovich
- Children's Health Research Center, Sanford Research, Sioux FallsSouth DakotaUSA
- Department of PediatricsThe University of South Dakota Sanford School of MedicineVermillion, South DakotaUSA
| | - Jill M. Weimer
- Children's Health Research Center, Sanford Research, Sioux FallsSouth DakotaUSA
- Department of PediatricsThe University of South Dakota Sanford School of MedicineVermillion, South DakotaUSA
| | - Daniel J. Kota
- Children's Health Research Center, Sanford Research, Sioux FallsSouth DakotaUSA
| |
Collapse
|
10
|
Shamosi A, Farokhi M, Ai J, Sharifi E. Induction of spontaneous neo-angiogenesis and tube formation in human endometrial stem cells by bioglass. JOURNAL OF MEDICAL HYPOTHESES AND IDEAS 2015. [DOI: 10.1016/j.jmhi.2015.09.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
11
|
Ye L, Jiang WG. Bone morphogenetic proteins in tumour associated angiogenesis and implication in cancer therapies. Cancer Lett 2015; 380:586-597. [PMID: 26639195 DOI: 10.1016/j.canlet.2015.10.036] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 09/17/2015] [Accepted: 10/12/2015] [Indexed: 02/09/2023]
Abstract
Bone morphogenetic protein (BMP) belongs to transforming growth factor-β superfamily. To date, more than 20 BMPs have been identified in humans. BMPs play a critical role in embryonic and postnatal development, and also in maintaining homeostasis in different organs and tissues by regulating cell differentiation, proliferation, survival and motility. They play important roles in the development and progression of certain malignancies, including prostate cancer, breast cancer, lung cancer, etc. Recently, more evidence shows that BMPs are also involved in tumour associated angiogenesis. For example BMP can either directly regulate the functions of vascular endothelial cells or indirectly influence the angiogenesis via regulation of angiogenic factors, such as vascular endothelial growth factor (VEGF). Such crosstalk can also be reflected in the interaction with other angiogenic factors, like hepatocyte growth factor (HGF) and basic fibroblast growth factor (bFGF). All these factors are involved in the orchestration of the angiogenic process during tumour development and progression. Review of the relevant studies will provide a comprehensive prospective on current understanding and shed light on the corresponding therapeutic opportunity.
Collapse
Affiliation(s)
- Lin Ye
- Metastasis & Angiogenesis Research Group, Cardiff University-Peking University Cancer Institute, Institute of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, UK.
| | - Wen G Jiang
- Metastasis & Angiogenesis Research Group, Cardiff University-Peking University Cancer Institute, Institute of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, UK
| |
Collapse
|
12
|
Altered chemokine signalling in endothelial progenitor cells from acute ulcerative colitis patients. Gastroenterol Res Pract 2015; 2015:843980. [PMID: 25737719 PMCID: PMC4337053 DOI: 10.1155/2015/843980] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Accepted: 01/25/2015] [Indexed: 12/18/2022] Open
Abstract
Ulcerative colitis (UC) is a chronic, idiopathic, inflammatory bowel disease, characterized by alternating stages of clinically active and inactive disease. UC exhibits several inflammatory characteristics, including immune activation, leukocyte infiltration, and altered vascular density. In UC, many of the upregulated inflammatory cytokines are proangiogenic and are released by diverse cell populations, such as infiltrating immune cells and endothelial cells (EC). Increasing evidences suggest that neovascularisation may involve also endothelial progenitor cells (EPCs). In this study we evaluated EPCs recruitment and homing, assessed by CXCR4 expression, in both acute and remitting phase of UC. We report an overall decrease of EPCs in UC patients (controls = 97,94 ± 37,34 cells/mL; acute = 31,10 ± 25,38 cells/mL; remitting = 30,33 ± 19,02 cells/mL; P < 0.001 for both UC groups versus controls). Moreover CXCR4+-EPCs, committed to home in inflammatory conditions, were found to be reduced in acute UC patients compared to both remitting patients and controls (acute = 3,13 ± 4,61 cells/mL; controls = 20,12 ± 14,0; remitting = 19,47 ± 12,83; P < 0,001). Interestingly, we found that administration of anti-inflammatory drugs in acute UC is associated with an increase in circulating EPCs, suggesting that this therapy may exert a strong influence on the progenitor cells response to inflammatory processes.
Collapse
|
13
|
Shi H, Jiang J, Ji J, Shi M, Cai Q, Chen X, Yu Y, Liu B, Zhu Z, Zhang J. Anti-angiogenesis participates in antitumor effects of metronomic capecitabine on colon cancer. Cancer Lett 2014; 349:128-35. [PMID: 24746899 DOI: 10.1016/j.canlet.2014.04.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 03/22/2014] [Accepted: 04/06/2014] [Indexed: 12/22/2022]
|
14
|
Ostojic A, Crowe S, McNeill B, Ruel M, Suuronen EJ. Preparation and characterization of circulating angiogenic cells for tissue engineering applications. Methods Mol Biol 2014; 1181:27-38. [PMID: 25070324 DOI: 10.1007/978-1-4939-1047-2_3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Circulating angiogenic cells (CACs) are a heterogeneous cell population of bone marrow (BM) origin. These cells are most commonly derived from the peripheral blood, bone marrow, and cord blood, and are one of the leading candidates for promoting vascularization in tissue engineering therapies. CACs can be isolated by culturing peripheral blood mononuclear cells (PBMCs) on fibronectin or by flow cytometry to obtain more specific subpopulations. Here we will describe how to generate a population of CACs, and how to characterize the cells and confirm their phenotype. Also, we will provide select methods that can be used to assess the angiogenic and endothelial cell-like properties of the CACs.
Collapse
Affiliation(s)
- Aleksandra Ostojic
- Division of Cardiac Surgery, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON, Canada, K1Y 4W7
| | | | | | | | | |
Collapse
|
15
|
Traish AM, Galoosian A. Androgens modulate endothelial function and endothelial progenitor cells in erectile physiology. Korean J Urol 2013; 54:721-31. [PMID: 24255752 PMCID: PMC3830963 DOI: 10.4111/kju.2013.54.11.721] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 09/24/2013] [Indexed: 12/21/2022] Open
Abstract
The incidence of erectile dysfunction (ED) increases with age and cardiovascular disease risk factors, such as hypertension, hyperlipidemia, insulin resistance, obesity, and diabetes. These risk factors are thought to contribute to endothelial dysfunction and atherosclerosis, thus contributing to the pathophysiology of ED. The role of the endothelium in regulating erectile physiology is well established. However, the role of androgens in modulating endothelial function and endothelial repair mechanisms subsequent to vascular injury in erectile tissue remains a subject of intensive research. The clinical and preclinical evidence discussed in this review suggests that androgens regulate endothelial function and also play an important role in the development and maturation of endothelial progenitor cells (EPCs), which are thought to play a critical role in repair of endothelial injury in vascular beds. In this review, we discuss the data available on the effects of androgens on endothelial function and EPCs in the repair of vascular injury. Indeed, more research is needed to fully understand the molecular and cellular basis of androgen action in regulating the development, differentiation, maturation, migration, and homing of EPCs to the site of injury. A better understanding of these processes will be critical to the development of new therapeutic approaches to the treatment of vascular ED.
Collapse
Affiliation(s)
- Abdulmaged M Traish
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA. ; Department of Urology, Boston University School of Medicine, Boston, MA, USA
| | | |
Collapse
|
16
|
Kim YB, Chung YW, Bae HS, Lee JK, Lee NW, Lee KW, Song JY. Circulating endothelial progenitor cells in gynaecological cancer. J Int Med Res 2013; 41:293-9. [DOI: 10.1177/0300060513476999] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Objectives To compare the frequency and absolute numbers of circulating endothelial progenitor cells (EPCs) in healthy control subjects and patients with gynaecological cancer, and to test the hypothesis that cancer treatment lowers EPC numbers. Methods Patients with cervical or ovarian cancer and healthy control subjects provided peripheral blood samples for the isolation of mononuclear cells. EPCs were identified by quadruple immunofluorescence staining and flow cytometry as CD45–/CD34+/CD133+/vascular endothelial growth factor receptor 2 (VEGFR2)+ cells. Results In total, 28 participants were enrolled. Circulating EPCs were present at higher frequencies (and in greater absolute numbers) in patients with cervical or ovarian cancer ( n = 14) than in controls ( n = 14). Concurrent chemoradiation therapy or surgery significantly reduced the frequency and number of EPCs in patients with gynaecological cancer, compared with pretreatment levels. Conclusions EPC levels decline throughout cancer treatment; their measurement may therefore be a useful surrogate marker to monitor treatment response.
Collapse
Affiliation(s)
- Yoon Byoung Kim
- Department of Obstetrics and Gynaecology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Ye Won Chung
- Department of Obstetrics and Gynaecology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hyo Sook Bae
- Department of Obstetrics and Gynaecology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jae Kwan Lee
- Department of Obstetrics and Gynaecology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Nak Woo Lee
- Department of Obstetrics and Gynaecology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Kyu Wan Lee
- Department of Obstetrics and Gynaecology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jae Yun Song
- Department of Obstetrics and Gynaecology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
17
|
Decrease in circulating endothelial progenitor cells in treated glioma patients. J Neurooncol 2012; 108:123-9. [DOI: 10.1007/s11060-012-0805-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Accepted: 01/25/2012] [Indexed: 01/10/2023]
|
18
|
DuBois SG, Stempak D, Wu B, Mokhtari RB, Nayar R, Janeway KA, Goldsby R, Grier HE, Baruchel S. Circulating endothelial cells and circulating endothelial precursor cells in patients with osteosarcoma. Pediatr Blood Cancer 2012; 58:181-4. [PMID: 21319292 PMCID: PMC3070958 DOI: 10.1002/pbc.23046] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2010] [Accepted: 12/29/2010] [Indexed: 02/03/2023]
Abstract
BACKGROUND Circulating endothelial cells (CECs) have been detected at increased numbers in patients with solid cancers. CECs have not been systematically evaluated in patients with osteosarcoma. PROCEDURE Patients 12 months to 30 years of age with newly diagnosed high-grade osteosarcoma were eligible for this prospective cohort study. Patients provided a single blood sample at study entry for CEC quantification by flow cytometry at a single reference laboratory. CECs were defined as CD146+, CD31+, CD45-, and CD133-. CEC progenitor cells (CEPs) were defined as CD146+, CD31+, CD45-, and CD133+. RESULTS Eighteen patients enrolled (11 males; median age 16 years; range 5-21 years). CEC counts did not differ between patients with osteosarcoma compared to seven pediatric healthy controls (median 645 cells/ml, range 60-5,320 cells/ml vs. 1,670 cells/ml, range 330-4,700 cells/ml, respectively; P = 0.12). CEP counts did not differ between patients compared to controls (median 126 cells/ml, range 0-5,320 cells/ml vs. median 260 cells/ml, range 0-10,670 cells/ml, respectively; P = 0.69). CEC and CEP counts did not correlate with metastatic status, tumor size, or histologic response to neoadjuvant chemotherapy. CONCLUSIONS CEC and CEP levels are not increased in patients with osteosarcoma compared to healthy controls. CECs and CEPs do not correlate with clinical features of osteosarcoma. Alternative novel markers of disease burden and response are needed in this disease.
Collapse
Affiliation(s)
- Steven G. DuBois
- Department of Pediatrics, UCSF School of Medicine, San Francisco, CA (SGD and RG)
| | - Diana Stempak
- Department of Hematology/Oncology, Hospital for Sick Children (DS, BW, RM, and SB)
| | - Bing Wu
- Department of Hematology/Oncology, Hospital for Sick Children (DS, BW, RM, and SB)
| | - Reza Bayat Mokhtari
- Department of Hematology/Oncology, Hospital for Sick Children (DS, BW, RM, and SB)
| | - Rakesh Nayar
- Princess Margaret Hospital, Toronto, Canada (RN)
| | - Katherine A. Janeway
- Department of Pediatrics, Dana-Farber Cancer Institute, Children's Hospital Boston, and Harvard Medical School, Boston, MA (KAJ and HEG)
| | - Robert Goldsby
- Department of Pediatrics, UCSF School of Medicine, San Francisco, CA (SGD and RG)
| | - Holcombe E. Grier
- Department of Pediatrics, Dana-Farber Cancer Institute, Children's Hospital Boston, and Harvard Medical School, Boston, MA (KAJ and HEG)
| | - Sylvain Baruchel
- Department of Hematology/Oncology, Hospital for Sick Children (DS, BW, RM, and SB)
| |
Collapse
|
19
|
Rae PC, Kelly RDW, Egginton S, St John JC. Angiogenic potential of endothelial progenitor cells and embryonic stem cells. Vasc Cell 2011; 3:11. [PMID: 21569302 PMCID: PMC3108917 DOI: 10.1186/2045-824x-3-11] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Accepted: 05/11/2011] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Endothelial progenitor cells (EPCs) are implicated in a range of pathological conditions, suggesting a natural therapeutic role for EPCs in angiogenesis. However, current angiogenic therapies involving EPC transplantation are inefficient due to rejection of donor EPCs. One solution is to derive an expanded population of EPCs from stem cells in vitro, to be re-introduced as a therapeutic transplant. To demonstrate the therapeutic potential of EPCs we performed in vitro transplantation of EPCs into endothelial cell (EC) tubules using a gel-based tubule formation assay. We also described the production of highly angiogenic EPC-comparable cells from pluripotent embryonic stem cells (ESCs) by direct differentiation using EC-conditioned medium (ECCM). RESULTS The effect on tubule complexity and longevity varied with transplantation quantity: significant effects were observed when tubules were transplanted with a quantity of EPCs equivalent to 50% of the number of ECs originally seeded on to the assay gel but not with 10% EPC transplantation. Gene expression of the endothelial markers VEGFR2, VE-cadherin and CD31, determined by qPCR, also changed dynamically during transplantation. ECCM-treated ESC-derived progenitor cells exhibited angiogenic potential, demonstrated by in vitro tubule formation, and endothelial-specific gene expression equivalent to natural EPCs. CONCLUSIONS We concluded the effect of EPCs is cumulative and beneficial, relying on upregulation of the angiogenic activity of transplanted cells combined with an increase in proliferative cell number to produce significant effects upon transplantation. Furthermore, EPCs derived from ESCs may be developed for use as a rapidly-expandable alternative for angiogenic transplantation therapy.
Collapse
Affiliation(s)
- Peter C Rae
- Centre for Cardiovascular Sciences, College of Medical & Dental Sciences, University of Birmingham, UK
- Clinical Sciences Research Institute, Warwick Medical School, University of Warwick, UK
| | - Richard DW Kelly
- Clinical Sciences Research Institute, Warwick Medical School, University of Warwick, UK
- Centre for Reproduction & Development, Monash Institute of Medical Research, Clayton VIC 3168, Australia
| | - Stuart Egginton
- Centre for Cardiovascular Sciences, College of Medical & Dental Sciences, University of Birmingham, UK
| | - Justin C St John
- Clinical Sciences Research Institute, Warwick Medical School, University of Warwick, UK
- Centre for Reproduction & Development, Monash Institute of Medical Research, Clayton VIC 3168, Australia
| |
Collapse
|
20
|
Gökçe A, Demirtas A, Halis F, Ekmekcioglu O. The effects of phosphodiesterase type 5 inhibitors on penile rigidity variables during a period with no sexual stimulation: a laboratory setting double-blind study. BJU Int 2011; 107:264-7. [PMID: 20438562 DOI: 10.1111/j.1464-410x.2010.09390.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To investigate the effects of phosphodiesterase type 5 (PDE5) inhibitors on erectile variables during a period with no sexual stimulation in a laboratory setting double-blind study. PATIENTS AND METHODS In all, 80 men without erectile dysfunction (ED) but with lifelong premature ejaculation (PE) were included in the study. The men were divided equally in to four groups and received either placebo, vardenafil (10 mg), sildenafil (50 mg) or tadalafil (20 mg) in a double-blind study design. The men attended the laboratory following 3 days of sexual abstinence and placebo or one of the PDE5 inhibitors was ingested after ≥ 2 h of fasting and non-smoking. The men were then immediately placed in a silent room and real-time penile rigidity and tumescence monitoring with Rigiscan Plus (Rigiscan Plus® System, Osbon Medical Systems, Augusta, GA, USA) began. The men read some magazines or newspapers that contained no sexually stimulating material for 1.5 h. There was no interaction between the men and observer during the test period. Times to first measured and total durations of base and tip rigidities, and also total and per minute rigidity were evaluated. RESULTS The recorded base and/or tip rigidity ratios were 40% (eight of 20), 71% (12/17), 47% (nine of 19) and 70% (14/20) in men who took placebo, sildenafil, tadalafil and vardenafil, respectively (P = 0.126). The ratio of men who could obtain ≥ 60% base and/or tip rigidities were 10% (two of 20), 41% (seven of 17), 26% (five of 19) and 55% (11/20) in placebo, sildenafil, tadalafil and vardenafil groups, respectively (P < 0.05). The median time to first measured base rigidity was 58.0, 21.5, 54.5 and 57 min with placebo, sildenafil, tadalafil and vardenafil, respectively (P = 0032). The median total duration of recorded base rigidity was 4.0, 27.5, 10.0 and 11.5 min in men who took placebo, sildenafil, tadalafil and vardenafil, respectively (P = 0.013). The median total base rigidity (area under the curve) was 72.8, 699.0, 360.5 and 553.0 with placebo, sildenafil, tadalafil and vardenafil, respectively (P = 0.016). CONCLUSIONS Significant penile rigidities were obtained with PDE5 inhibitors during the short test period, with no sexual stimulation, in laboratory conditions. This finding might support the use of PDE5 inhibitors in men who need penile rehabilitation.
Collapse
Affiliation(s)
- Ahmet Gökçe
- Department of Urology, Erciyes University Medical Faculty, Kayseri, Turkey.
| | | | | | | |
Collapse
|
21
|
Yates CC, Krishna P, Whaley D, Bodnar R, Turner T, Wells A. Lack of CXC chemokine receptor 3 signaling leads to hypertrophic and hypercellular scarring. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 176:1743-55. [PMID: 20203286 DOI: 10.2353/ajpath.2010.090564] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
CXC chemokine receptor 3 (CXCR3) signaling promotes keratinocyte migration while terminating fibroblast and endothelial cell immigration into wounds; this signaling also directs epidermal and matrix maturation. Herein, we investigated the long-term effects of failure to activate the "stop-healing" CXCR3 axis. Full-thickness excisional wounds were created on CXCR3 knockout((-/-)) or wild-type mice and examined at up to 180 days after wounding. Grossly, the CXCR3(-/-) mice presented a thick keratinized scar compared with the wild-type mice in which the scar was scarcely noticeable; histological examination revealed thickening of both the epidermis and dermis. The dermis was disorganized with thick and long collagen fibrils and contained excessive collagen content in comparison with the wild-type mice. Interestingly, the CXCR3(-/-) wounds presented lower tensile/burst strength, which correlates with decreased alignment of collagen fibers, similar to published findings of human scars. Persistent Extracellular matrix turnover and immaturity was shown by the elevated expression of proteins of the immature matrix as well as expression of matrix metallopeptidase-9 MMP-9. Interestingly, the scars in the CXCR3(-/-) mice presented evidence of de novo development of a sterile inflammatory response only months after wounding; earlier periods showed resolution of the initial inflammatory stage. These in vivo studies establish that the absence of CXCR3(-/-) signaling network results in hypertrophic and hypercellular scarring characterized by on-going wound regeneration, cellular proliferation, and scars in which immature matrix components are undergoing increased turnover resulting in a chronic inflammatory process.
Collapse
Affiliation(s)
- Cecelia C Yates
- University of Pittsburgh, Department of Pathology, Pittsburgh, PA 15261, USA
| | | | | | | | | | | |
Collapse
|
22
|
Cicco G, Cicco S. The Influence of Oxygen Supply, Hemorheology and Microcirculation in the Heart and Vascular Systems. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 662:33-9. [DOI: 10.1007/978-1-4419-1241-1_4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
23
|
Ran S, Volk L, Hall K, Flister MJ. Lymphangiogenesis and lymphatic metastasis in breast cancer. ACTA ACUST UNITED AC 2009; 17:229-51. [PMID: 20036110 DOI: 10.1016/j.pathophys.2009.11.003] [Citation(s) in RCA: 181] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2009] [Revised: 04/11/2009] [Accepted: 10/23/2009] [Indexed: 01/03/2023]
Abstract
Lymphatic metastasis is the main prognostic factor for survival of patients with breast cancer and other epithelial malignancies. Mounting clinical and experimental data suggest that migration of tumor cells into the lymph nodes is greatly facilitated by lymphangiogenesis, a process that generates new lymphatic vessels from pre-existing lymphatics with the aid of circulating lymphatic endothelial progenitor cells. The key protein that induces lymphangiogenesis is vascular endothelial growth factor receptor-3 (VEGFR-3), which is activated by vascular endothelial growth factor-C and -D (VEGF-C and VEGF-D). These lymphangiogenic factors are commonly expressed in malignant, tumor-infiltrating and stromal cells, creating a favorable environment for generation of new lymphatic vessels. Clinical evidence demonstrates that increased lymphatic vessel density in and around tumors is associated with lymphatic metastasis and reduced patient survival. Recent evidence shows that breast cancers induce remodeling of the local lymphatic vessels and the regional lymphatic network in the sentinel and distal lymph nodes. These changes include an increase in number and diameter of tumor-draining lymphatic vessels. Consequently, lymph flow away from the tumor is increased, which significantly increases tumor cell metastasis to draining lymph nodes and may contribute to systemic spread. Collectively, recent advances in the biology of tumor-induced lymphangiogenesis suggest that chemical inhibitors of this process may be an attractive target for inhibiting tumor metastasis and cancer-related death. Nevertheless, this is a relatively new field of study and much remains to be established before the concept of tumor-induced lymphangiogenesis is accepted as a viable anti-metastatic target. This review summarizes the current concepts related to breast cancer lymphangiogenesis and lymphatic metastasis while highlighting controversies and unanswered questions.
Collapse
Affiliation(s)
- Sophia Ran
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, 801 N. Rutledge, Springfield, IL 62794-9678, USA
| | | | | | | |
Collapse
|
24
|
Hofman P. [Circulating tumor cells: pitfalls and new promises in clinical oncology]. Ann Pathol 2009; 29 Spec No 1:S25-7. [PMID: 19887244 DOI: 10.1016/j.annpat.2009.07.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2009] [Accepted: 07/26/2009] [Indexed: 10/20/2022]
Affiliation(s)
- Paul Hofman
- Inserm ERI-21/EA 4319, laboratoire de pathologie clinique et expérimentale, hôpital Pasteur, biobanque CHUN, faculté de médecine de Nice, université de Nice-Sophia-Antipolis, avenue de la voie-romaine-hôpital-Pasteur, BP 69, Nice, France.
| |
Collapse
|
25
|
Goldstein MR, Mascitelli L, Pezzetta F. Primary prevention of cardiovascular disease with statins: cautionary notes. QJM 2009; 102:817-20. [PMID: 19620217 DOI: 10.1093/qjmed/hcp099] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
26
|
Garolla A, D'Incà R, Checchin D, Biagioli A, De Toni L, Nicoletti V, Scarpa M, Bolzonello E, Sturniolo GC, Foresta C. Reduced endothelial progenitor cell number and function in inflammatory bowel disease: a possible link to the pathogenesis. Am J Gastroenterol 2009; 104:2500-7. [PMID: 19568231 DOI: 10.1038/ajg.2009.332] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Circulating endothelial progenitor cells (EPCs) are essential for endothelial repair and vascular healing. Patients with inflammatory bowel disease (IBD) may suffer from endothelial dysfunction. Reduced EPC number, impaired mobilization, or increased EPC apoptosis may be crucial in this phenomenon. The aim of our study was to investigate the number and function of EPCs in patients with IBD and to assess their endothelial function. METHODS In 100 IBD patients (47 ulcerative colitis (UC) and 53 Crohn's disease (CD)) and 50 healthy controls, EPC number, CXC motif receptor 4 (CXCR4) expression, the percentage of apoptotic circulating EPCs, and the number of colony-forming units were evaluated. Endothelial dysfunction was assessed by luteinizing hormone (LH), follicle stimulating hormone (FSH), and testosterone levels, and in a subgroup of patients, brachial artery flow-mediated dilation (FMD) was measured. Kruskal-Wallis ANOVA (analysis of variance), Mann-Whitney U two-tailed, and Spearman's rank correlation tests were used to assess differences. RESULTS EPC number was significantly lower in UC patients (39.6 (95% confidence interval (95% CI): 30.7-48.6)) and in CD patients (43.1 (95% CI: 35.9-50.4)) than in healthy controls (97.1 (95% CI: 88.3-105.9)), (P<0.001). LH and FSH levels and CXCR4 expression on EPCs did not significantly differ from controls. Testosterone concentrations and FMD were lower in UC patients. Number of apoptotic EPCs was higher in both UC and CD patients with an impaired ability to generate colony in vitro. CONCLUSIONS We hypothesize that in IBD patients, apoptosis contributes to the reduction of circulating EPC number and to their ability to proliferate in vitro. As this condition represents a risk factor for cardiovascular disease, endothelial function should be evaluated in these patients.
Collapse
Affiliation(s)
- Andrea Garolla
- Department of Histology, Microbiology, and Medical Biotechnologies, Center for Male Gamete Cryopreservation, University of Padova, Padova, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Francavilla C, Maddaluno L, Cavallaro U. The functional role of cell adhesion molecules in tumor angiogenesis. Semin Cancer Biol 2009; 19:298-309. [DOI: 10.1016/j.semcancer.2009.05.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2009] [Accepted: 05/20/2009] [Indexed: 12/18/2022]
|
28
|
Hartman J, Ström A, Gustafsson JA. Estrogen receptor beta in breast cancer--diagnostic and therapeutic implications. Steroids 2009; 74:635-41. [PMID: 19463683 DOI: 10.1016/j.steroids.2009.02.005] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2008] [Revised: 02/17/2009] [Accepted: 02/18/2009] [Indexed: 02/07/2023]
Abstract
More than 10 years have passed since the discovery of the second estrogen receptor, estrogen receptor beta (ERbeta). It is now evident that ERalpha is not the only ER in breast cancer cells; in fact, ERbeta is expressed in the majority of breast cancers although at lower levels than in the normal breast. In addition, ERbeta is expressed in breast cancer infiltrating lymphocytes, fibroblasts and endothelial cells, all known to influence tumor growth. By overexpressing or knocking-out ERbeta in breast cancer cell lines, several researchers have investigated its function with respect to proliferation and tumor growth. It appears that ERbeta is anti-proliferative, in many ways antagonising the function of ERalpha. Furthermore, phytoestrogens have a binding-preference for ERbeta and several epidemiological studies indicate a breast cancer preventing effect of this class of compounds. Tamoxifen is one of the standard, adjuvant treatments for ERalpha positive breast cancer, classically thought to mediate its effect through ERalpha. However, in several recent studies, ERbeta has been described as a potential marker for tamoxifen response. In summary, experimental, epidemiological as well as diagnostic studies point towards ERbeta as an important factor in breast cancer, opening up the possibility for novel ERbeta-selective therapies in the treatment of breast cancer.
Collapse
Affiliation(s)
- Johan Hartman
- Department of Biosciences and Nutrition, Novum, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | | | | |
Collapse
|
29
|
Alghisi GC, Rüegg C. Vascular Integrins in Tumor Angiogenesis: Mediators and Therapeutic Targets. ACTA ACUST UNITED AC 2009; 13:113-35. [PMID: 16728329 DOI: 10.1080/10623320600698037] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The notion that tumor angiogenesis may have therapeutic implications in the control of tumor growth was introduced by Dr. Judah Folkman in 1971. The approval of Avastin in 2004 as the first antiangiogenic systemic drug to treat cancer patients came as a validation of this visionary concept and opened new perspectives to the treatment of cancer. In addition, this success boosted the field to the quest for new therapeutic targets and antiangiogenic drugs. Preclinical and clinical evidence indicate that vascular integrins may be valid therapeutic targets. In preclinical studies, pharmacological inhibition of integrin function efficiently suppressed angiogenesis and inhibited tumor progression. alphaVbeta3 and alphaVbeta5 were the first vascular integrins targeted to suppress tumor angiogenesis. Subsequent experiments revealed that at least four additional integrins (i.e., alpha1beta1, alpha2beta1, alpha5beta1, and alpha6beta4) might be potential therapeutic targets. In clinical studies low-molecular-weight integrin inhibitors and anti-integrin function-blocking antibodies demonstrated low toxicity and good tolerability and are now being tested in combination with radiotherapy and chemotherapy for anticancer activity in patients. In this article the authors review the role of integrins in angiogenesis, present recent development in the use of alphaVbeta3 and alpha5beta1 integrin antagonists as potential therapeutics in cancer, and discuss future perspectives.
Collapse
Affiliation(s)
- Gian Carlo Alghisi
- Centre Pluridisciplinaire d'Oncologie (CePO), Faculty of Biology and Medicine, University of Lausanne, Switzerland
| | | |
Collapse
|
30
|
Taketo MM. Role of bone marrow-derived cells in colon cancer: lessons from mouse model studies. J Gastroenterol 2009; 44:93-102. [PMID: 19214670 DOI: 10.1007/s00535-008-2321-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2008] [Accepted: 11/04/2008] [Indexed: 02/04/2023]
Abstract
The role of the tumor stroma in carcinogenesis and cancer progression have been documented for a long time. However, the molecules and mechanisms involved have not been understood precisely. Recently, various mediators involved in the communication between the tumor epithelium and stroma and their roles have been revealed by utilizing new technology such as array analysis, laser capture sampling, and genetically altered mice. Moreover, accumulating evidence indicates that some cells in the tumor stroma are derived from the bone marrow (BM). While some of these BM-derived cells are well-known players in inflammation, as exemplified by macrophages, other types of BM-derived cells have been described only recently and are still poorly characterized. In this review, I focus on the latter class of BM-derived cells in colon carcinogenesis, with reference to similar cells in other types of cancer as well. Studies of these myeloid cells should help us understand the inflammation and immune response from a broader perspective as the body's reaction to pathogenic insults.
Collapse
Affiliation(s)
- Makoto Mark Taketo
- Department of Pharmacology, Graduate School of Medicine, Kyoto University, Yoshida-Konoé-cho, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
31
|
Abstract
Recent studies have found that bone marrow-derived cells give rise to endothelial cells during states of tissue repair and disease. We have found that one key integrin, integrin-alpha4beta1, promotes the homing of circulating endothelial progenitor cells (EPCs) to sites of ongoing tissue repair. This integrin facilitates the adhesion of EPCs to the vascular endothelium in inflamed tissue or within tumors. We demonstrate how to identify, isolate, purify, and characterize EPCs. We also demonstrate in vivo analysis of the roles of bone marrow-derived cells in tumor growth and angiogenesis by demonstrating adoptive transfer, bone marrow transplantation, tumor models, and immunohistochemistry for markers of blood and endothelial vessels. Finally, we show how to characterize cell adhesion mechanisms regulating bone marrow-derived progenitor cell trafficking.
Collapse
|
32
|
Abstract
The extracellular matrix (ECM) acts both as a physical scaffold for cells and as a repository for growth factors. Moreover, ECM structure and physical-chemical properties convey precise information to cells that profoundly influences their biology by interactions with cell surface receptors termed integrins. During angiogenesis, the perivascular ECM plays a critical role in determining the proliferative, invasive and survival responses of the local vascular cells to the angiogenic growth factors. Dynamic changes in both the ECM and the local vascular cells act in concert to regulate new blood vessel growth. The digestion of ECM components by proteolysis is critical for the invasive capacity of endothelial cells, but also creates ECM fragments, which antagonize the mechanosensory function of integrins, and can be apoptogenic. Here, we discuss the roles of integrins in modulating cellular responses to a changing ECM, in particular the regulation of survival and invasion among invasive endothelial cells.
Collapse
|
33
|
Iohara K, Zheng L, Wake H, Ito M, Nabekura J, Wakita H, Nakamura H, Into T, Matsushita K, Nakashima M. A novel stem cell source for vasculogenesis in ischemia: subfraction of side population cells from dental pulp. Stem Cells 2008; 26:2408-18. [PMID: 18583536 DOI: 10.1634/stemcells.2008-0393] [Citation(s) in RCA: 176] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cell therapy with stem cells and endothelial progenitor cells (EPCs) to stimulate vasculogenesis as a potential treatment for ischemic disease is an exciting area of research in regenerative medicine. EPCs are present in bone marrow, peripheral blood, and adipose tissue. Autologous EPCs, however, are obtained by invasive biopsy, a potentially painful procedure. An alternative approach is proposed in this investigation. Permanent and deciduous pulp tissue is easily available from teeth after extraction without ethical issues and has potential for clinical use. We isolated a highly vasculogenic subfraction of side population (SP) cells based on CD31 and CD146, from dental pulp. The CD31(-);CD146(-) SP cells, demonstrating CD34+ and vascular endothelial growth factor-2 (VEGFR2)/Flk1+, were similar to EPCs. These cells were distinct from the hematopoietic lineage as CD11b, CD14, and CD45 mRNA were not expressed. They showed high proliferation and migration activities and multilineage differentiation potential including vasculogenic potential. In models of mouse hind limb ischemia, local transplantation of this subfraction of SP cells resulted in successful engraftment and an increase in the blood flow including high density of capillary formation. The transplanted cells were in proximity of the newly formed vasculature and expressed several proangiogenic factors, such as VEGF-A, G-CSF, GM-CSF, and MMP3. Conditioned medium from this subfraction showed the mitogenic and antiapoptotic activity on human umbilical vein endothelial cells. In conclusion, subfraction of SP cells from dental pulp is a new stem cell source for cell-based therapy to stimulate angiogenesis/vasculogenesis during tissue regeneration.
Collapse
Affiliation(s)
- Koichiro Iohara
- Department of Oral Disease Research, National Institute for Longevity Sciences, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
|
35
|
Scott RC, Crabbe D, Krynska B, Ansari R, Kiani MF. Aiming for the heart: targeted delivery of drugs to diseased cardiac tissue. Expert Opin Drug Deliv 2008; 5:459-70. [DOI: 10.1517/17425247.5.4.459] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
36
|
Spannuth WA, Sood AK, Coleman RL. Angiogenesis as a strategic target for ovarian cancer therapy. ACTA ACUST UNITED AC 2008; 5:194-204. [PMID: 18268546 DOI: 10.1038/ncponc1051] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2007] [Accepted: 10/22/2007] [Indexed: 01/07/2023]
Abstract
Angiogenesis is a complex and highly regulated process that is crucial for tumor growth and metastasis. Insights into the molecular mechanisms of tumor angiogenesis have led to the identification of potential angiogenic targets and the development of novel antivascular agents. Many of these agents are being evaluated in clinical trials and have shown promising antitumor activity. This Review highlights the results of the latest clinical studies of antivascular agents in ovarian cancer and discusses the challenges and opportunities for future clinical trials.
Collapse
Affiliation(s)
- Whitney A Spannuth
- Departments of Gynecologic Oncology and Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas 77230-1439, USA
| | | | | |
Collapse
|
37
|
Salehi F, Kovacs K, Cusimano MD, Horvath E, Bell CD, Rotondo F, Scheithauer BW. Immunohistochemical expression of nestin in adenohypophysial vessels during development of pituitary infarction. J Neurosurg 2008; 108:118-23. [PMID: 18173320 DOI: 10.3171/jns/2008/108/01/0118] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVES The aim of this work was to investigate the immunohistochemical expression of nestin, a member of the intermediate filament family, in adenohypophysial vasculature during development and progression of pituitary infarction. METHODS Forty-five nontumorous adenohypophyses and 34 pituitary adenomas of various types, all exhibiting acute or healing infarcts, were examined immunohistochemically using the streptavidin-biotin-peroxidase complex method. RESULTS In both adenohypophyses and pituitary adenomas without infarction, nestin was expressed in only a few capillaries and endothelial cells. In acute infarcts without a vascular response, no nestin was demonstrable within necrotic capillaries (50 cases). In organizing infarcts, newly formed vessels spreading into necrotic zones showed nestin expression in all capillaries and practically every endothelial cell (25 cases). In the hypocellular, fibrotic scar phase, only a few vessels (4) were apparent, and immunoreactivity was focal and mild. CONCLUSIONS Nestin is strongly expressed in newly formed capillaries and is downregulated when infarcts transform to fibrous tissue. Nestin expression may provide valuable insight into the process of pituitary angiogenesis.
Collapse
Affiliation(s)
- Fateme Salehi
- Department of Laboratory Medicine, St. Michael's Hospital, University of Toronto, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
38
|
Foresta C, Zuccarello D, De Toni L, Garolla A, Caretta N, Ferlin A. Androgens stimulate endothelial progenitor cells through an androgen receptor-mediated pathway. Clin Endocrinol (Oxf) 2008; 68:284-9. [PMID: 17803706 DOI: 10.1111/j.1365-2265.2007.03036.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
BACKGROUND AND OBJECTIVE Testosterone (T) treatment has recently been shown to induce an increase in the number of endothelial progenitor cells (EPCs) through a possible effect on bone marrow. Hypogonadotrophic hypogonadal (HH) men have low circulating EPCs that increase significantly after T treatment. Moreover, expression of the androgen receptor (AR) has been demonstrated by immunohistochemistry in these cells, suggesting that T might also have a direct effect on EPC function. In the present study we investigated the expression and function of the AR in human EPCs and the in vitro effect of androgens on EPC function. Design and patients EPCs obtained from healthy male anonymous blood donors were analysed after androgen stimulation with and without AR antagonist administration (flutamide). RESULTS Reverse transcriptase polymerase chain reaction (RT-PCR), Western blotting and immunofluorescence analyses demonstrated the expression of AR mRNA and protein in human EPCs. Stimulation of these cells with the synthetic androgen methyltrienolone (R1881) caused AR translocation in the nucleus, suggesting its activation. Colony forming unit (CFU), proliferation and migration assays under different doses of R1881 demonstrated a dose-dependent increase in EPC proliferation, migration and colony formation. All these effects are abolished by flutamide pretreatment. CONCLUSIONS This study showed that the increase in the proliferation, migration and colony formation activity of EPCs induced by androgens is an AR-mediated pathway. Androgen exerts these effects at concentrations that are physiologically present in men and therefore further studies are needed to clarify the clinical significance of these effects in normal and pathological conditions.
Collapse
Affiliation(s)
- Carlo Foresta
- Department of Histology, Microbiology and Medical Biotechnologies, Centre for Male Gamete Cryopreservation, University of Padova, Padova, Italy.
| | | | | | | | | | | |
Collapse
|
39
|
Nogueras S, Merino A, Ojeda R, Carracedo J, Rodriguez M, Martin-Malo A, Ramírez R, Aljama P. Coupling of endothelial injury and repair: an analysis using an in vivo experimental model. Am J Physiol Heart Circ Physiol 2008; 294:H708-13. [DOI: 10.1152/ajpheart.00466.2007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The repair of the endothelium after inflammatory injury is essential to maintaining homeostasis. The link between inflammation-induced endothelial damage and repair has not been fully characterized in vivo. We have developed a rat model to evaluate the coupling of lipopolysaccharide (LPS)-induced endothelial injury and repair. Aortic endothelium injury was analyzed by both inmunohistochemistry and flow cytometry to quantify the number of endothelial cells and the percentage of apoptotic endothelial cells. We have also identified the percentage of circulating angiogenic cells capable of repairing the damaged endothelium. Erythropoietin was administered to inhibit LPS-induced endothelial apoptosis. Loss of the normal endothelial structure was observed in the aorta of the animals treated with LPS. Eight hours after LPS administration, the number of endothelial cells decreased by 40%, returning to normal after 24 h. There was a threefold increase in the percentage of circulating angiogenic cells, which did not return to normal levels until 48 h after LPS administration. Circulating angiogenic cell levels did not change when LPS-induced endothelial damage was prevented by erythropoietin. The endothelial injury caused by inflammation activates the mobilization of circulating angiogenic cells, thus completing endothelial repair. Inflammation without endothelial injury does not trigger the mobilization of circulating angiogenic cells.
Collapse
|
40
|
Herdrich B, Lind R, Liechty K. Multipotent adult progenitor cells: their role in wound healing and the treatment of dermal wounds. Cytotherapy 2008; 10:543-50. [DOI: 10.1080/14653240802345820] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
41
|
Teloken PE, Mulhall JP. Impact of phosphodiesterase type 5 inhibitors on endothelial function. Rev Urol 2008; 10:26-30. [PMID: 18470271 PMCID: PMC2312340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
It is now known that endothelial health is essential for normal erectile function, and changes in endothelial integrity or function may lead to erectile dysfunction (ED). Because phosphodiesterase type 5 (PDE-5) inhibitors have been shown to improve endothelial function, many investigators have questioned whether PDE-5 inhibition will lead to improvement in erectile function. Data from the studies reviewed in this article show that therapy with PDE-5 inhibitors results in improvement in flow-mediated dilation, nocturnal penile tumescence and rigidity, and carotid artery intima-media thickness as well as higher scores on the Sexual Health Inventory for Men, International Index of Erectile Function, Erection Function Domain, and other instruments. Further research is needed to determine whether long-term PDE-5 inhibition can reverse ED and whether use of these agents will decrease cardiovascular morbidity in high-risk populations.
Collapse
Affiliation(s)
- Patrick E Teloken
- Department of Urology, Weill Medical College of Cornell University New York, NY
| | | |
Collapse
|
42
|
Badiavas EV, Ford D, Liu P, Kouttab N, Morgan J, Richards A, Maizel A. Long-term bone marrow culture and its clinical potential in chronic wound healing. Wound Repair Regen 2007; 15:856-65. [DOI: 10.1111/j.1524-475x.2007.00305.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
43
|
Di Fabio F, Alvarado C, Majdan A, Gologan A, Voda L, Mitmaker E, Beitel LK, Gordon PH, Trifiro M. Underexpression of mineralocorticoid receptor in colorectal carcinomas and association with VEGFR-2 overexpression. J Gastrointest Surg 2007; 11:1521-8. [PMID: 17703341 DOI: 10.1007/s11605-007-0234-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2007] [Accepted: 06/30/2007] [Indexed: 01/31/2023]
Abstract
BACKGROUND The human mineralocorticoid receptor (MR) is a steroid receptor widely expressed in colorectal mucosa. A significant role for the MR in the reduction of vascular endothelial growth factor receptor-2 (VEGFR-2) mRNA levels has been demonstrated in vitro. To evaluate a potential contribution of MR to colorectal carcinoma progression, we analyzed the expression of MR in relation to VEGFR-2. METHODS Fresh human colorectal cancer tissue and adjacent normal mucosa were harvested from 48 consecutive patients. MR and VEGFR-2 mRNA expression levels were determined by real-time reverse transcriptase-polymerase chain reaction and correlated with clinicopathological parameters. RESULTS A decline of MR expression was observed in all carcinomas compared to normal mucosa. Expression of MR was a median of 11-fold lower in carcinoma compared to the normal mucosa, irrespective of the location, size, stage, and differentiation. MR was a median of 20-fold underexpressed in carcinomas with VEGFR-2 overexpression vs only 9-fold in carcinomas with VEGFR-2 underexpression (p = 0.035, Mann-Whitney test). CONCLUSIONS These findings support the hypothesis that reduction of MR expression may be one of the early events involved in colorectal carcinoma progression. The inverse association between MR and VEGFR-2 expression in carcinoma suggests a potential tumor-suppressive function for MR.
Collapse
Affiliation(s)
- Francesco Di Fabio
- Department of Colon and Rectal Surgery, Sir Mortimer B. Davis Jewish General Hospital, Montreal, Quebec, Canada.
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
The growth of new blood vessels may be either beneficial or harmful. The angiogenic process may be measured by a variety of techniques, although it may often be the quality rather than quantity of resulting blood vessels that determines function. Endothelial cells play a key role in the initiation of angiogenesis, and vascular endothelial growth factor (VEGF) may be viewed as a prototypical direct-acting angiogenic factor. VEGF acts through multiple cell surface receptors and signaling pathways to stimulate endothelial cell proliferation, survival, and migration. By inducing other growth factor expression, VEGF stimulates a cascade of angiogenic activity. Different tissues may utilize various angiogenic pathways that are modulated by diverse host tissue responses. Furthermore, a single tissue may progress through a sequence of angiogenic pathways, for example, as acute injury progresses to chronic inflammation. The phenotype of the resulting neovasculature is critically dependent on the context in which it is formed. Biomarkers of angiogenesis are being developed as an aid to assessing human disease. Histological assessment of vascular density and angiogenic factor expression, in vivo imaging, Doppler ultrasound, and biofluid assays each may have clinical utility. Therapeutic targeting of angiogenesis will depend both on the generation of acceptable pharmacological agents and on the identification of patients who may and do gain benefit from such treatments.
Collapse
Affiliation(s)
- David A Walsh
- Academic Rheumatology, University of Nottingham Clinical Sciences Building, Nottingham City Hospital, Nottingham NG5 1PB, United Kingdom
| |
Collapse
|
45
|
Abstract
Since 1997, postnatal vasculogenesis has been purported to be an important mechanism for neoangiogenesis via bone marrow (BM)-derived circulating endothelial progenitor cells (EPCs). Based on this paradigm, EPCs have been extensively studied as biomarkers to assess severity of cardiovascular disease and as a cell-based therapy for several human cardiovascular disorders. In the majority of studies to date, EPCs were identified and enumerated by two primary methodologies; EPCs were obtained and quantified following in vitro cell culture, or EPCs were identified and enumerated by flow cytometry. Both methods have proven controversial. This review will attempt to outline the definition of EPCs from some of the most widely cited published reports in an effort to provide a framework for understanding subsequent studies in this rapidly evolving field. We will focus this review on studies that used cell culture techniques to define EPCs.
Collapse
Affiliation(s)
- D N Prater
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | | |
Collapse
|
46
|
Kitamura T, Kometani K, Hashida H, Matsunaga A, Miyoshi H, Hosogi H, Aoki M, Oshima M, Hattori M, Takabayashi A, Minato N, Taketo MM. SMAD4-deficient intestinal tumors recruit CCR1+ myeloid cells that promote invasion. Nat Genet 2007; 39:467-75. [PMID: 17369830 DOI: 10.1038/ng1997] [Citation(s) in RCA: 218] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2006] [Accepted: 02/12/2007] [Indexed: 12/16/2022]
Abstract
Inactivation of TGF-beta family signaling is implicated in colorectal tumor progression. Using cis-Apc(+/Delta716) Smad4(+/-) mutant mice (referred to as cis-Apc/Smad4), a model of invasive colorectal cancer in which TGF-beta family signaling is blocked, we show here that a new type of immature myeloid cell (iMC) is recruited from the bone marrow to the tumor invasion front. These CD34(+) iMCs express the matrix metalloproteinases MMP9 and MMP2 and the CC-chemokine receptor 1 (CCR1) and migrate toward the CCR1 ligand CCL9. In adenocarcinomas, expression of CCL9 is increased in the tumor epithelium. By deleting Ccr1 in the background of the cis-Apc/Smad4 mutant, we further show that lack of CCR1 prevents accumulation of CD34(+) iMCs at the invasion front and suppresses tumor invasion. These results indicate that loss of transforming growth factor-beta family signaling in tumor epithelium causes accumulation of iMCs that promote tumor invasion.
Collapse
Affiliation(s)
- Takanori Kitamura
- Department of Pharmacology, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Döme B, Hendrix MJC, Paku S, Tóvári J, Tímár J. Alternative vascularization mechanisms in cancer: Pathology and therapeutic implications. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 170:1-15. [PMID: 17200177 PMCID: PMC1762709 DOI: 10.2353/ajpath.2007.060302] [Citation(s) in RCA: 260] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Although cancer cells are not generally controlled by normal regulatory mechanisms, tumor growth is highly dependent on the supply of oxygen, nutrients, and host-derived regulators. It is now established that tumor vasculature is not necessarily derived from endothelial cell sprouting; instead, cancer tissue can acquire its vasculature by co-option of pre-existing vessels, intussusceptive microvascular growth, postnatal vasculogenesis, glomeruloid angiogenesis, or vasculogenic mimicry. The best-known molecular pathway driving tumor vascularization is the hypoxia-adaptation mechanism. However, a broad and diverse spectrum of genetic aberrations is associated with the development of the "angiogenic phenotype." Based on this knowledge, novel forms of antivascular modalities have been developed in the past decade. When applying these targeted therapies, the stage of tumor progression, the type of vascularization of the given cancer tissue, and the molecular machinery behind the vascularization process all need to be considered. A further challenge is finding the most appropriate combinations of antivascular therapies and standard radio- and chemotherapies. This review intends to integrate our recent knowledge in this field into a rational strategy that could be the basis for developing effective clinical modalities using antivascular therapy for cancer.
Collapse
Affiliation(s)
- Balázs Döme
- Department of Tumor Biology and Thoracic Oncology, National Koranyi Institute of Pulmonology, Budapest, Hungary
| | | | | | | | | |
Collapse
|
48
|
Futamura M, Kamino H, Miyamoto Y, Kitamura N, Nakamura Y, Ohnishi S, Masuda Y, Arakawa H. Possible role of semaphorin 3F, a candidate tumor suppressor gene at 3p21.3, in p53-regulated tumor angiogenesis suppression. Cancer Res 2007; 67:1451-60. [PMID: 17308083 DOI: 10.1158/0008-5472.can-06-2485] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although the regulation of tumor angiogenesis is believed to be one of the core functions of p53, the mechanism still remains to be elucidated. Here, we report that semaphorin 3F (SEMA3F), an axon guidance molecule, is involved in p53-regulated antiangiogenesis. The expression level of SEMA3F mRNA was increased by both exogenous and endogenous p53. Chromatin immunoprecipitation assay indicated that a potent p53-binding sequence in intron 1 of SEMA3F interacts with p53 and that it has a p53-responsive transcriptional activity. Overexpression of SEMA3F inhibited in vitro cell growth of the lung cancer cell line H1299. In nude mice assay, the size of the H1299 tumors expressing SEMA3F was much smaller, and they showed lesser number of blood vessels as compared with the control tumors. Moreover, tumors derived from the p53-knockdown colorectal cancer cell line LS174T displayed a remarkable enhancement of tumor vessel formation as compared with control tumors containing normal levels of p53. The expression levels of SEMA3F and neuropilin-2 (NRP2), the functional receptor for SEMA3F, in p53-knockdown LS174T tumors were lower than those in the control tumors. Adenovirus-mediated SEMA3F gene transfer induced the remarkable in vitro growth suppression of the stable transformant of H1299 cells, which express high levels of NRP2. These results suggest that p53 negatively regulates tumor vessel formation and cell growth via the SEMA3F-NRP2 pathway.
Collapse
MESH Headings
- Cell Growth Processes/genetics
- Cell Line, Tumor
- Chromosomes, Human, Pair 3
- Gene Expression Regulation, Neoplastic
- Genes, Tumor Suppressor
- Genes, p53
- HCT116 Cells
- Humans
- Membrane Proteins/biosynthesis
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Neoplasms/blood supply
- Neoplasms/genetics
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Nerve Tissue Proteins/biosynthesis
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Neuropilin-2/metabolism
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Signal Transduction
- Transcription, Genetic
Collapse
Affiliation(s)
- Manabu Futamura
- Cancer Medicine and Biophysics Division, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Angiogenesis (the growth of new blood vessels) is a complex multistep process that involves multiple cell types, numerous growth factors, and complex regulatory checks and balances. Tight control of vascular remodeling evolved to ensure stability of the vasculature while maintaining the body's ability to rapidly mount an angiogenic response requiring a high degree of plasticity. Angiogenesis is critical not only for physiological development, but also for the progression of pathologies, and is thus a target for therapeutic intervention. The importance of the process coupled with the ease of access for delivery of contrast agents makes the vasculature at large, and angiogenesis in particular, a favorable target of functional and molecular imaging. Recent developments in molecular imaging tools have expanded our views to encompass many components of the process. Functional imaging of blood volume, vessel permeability, and vasoreactivity is complemented by novel contrast agents that reveal specific targets on endothelial cells. Methods have been developed to label vascular cells so as to track their recruitment to sites of angiogenesis, and new "smart" contrast agents have been designed to reveal the activity of enzymatic reactions in altering the extracellular matrix (ECM) during angiogenesis.
Collapse
Affiliation(s)
- Michal Neeman
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel.
| | | | | | | |
Collapse
|
50
|
Abstract
This review considers the potential existence and role of stem or progenitor cell populations within the non-epithelial tissues of the oral mucosa. Currently, there is little published evidence supporting this hypothesis; however, because of the similarities in structure and function of the oral mucosa and skin, findings within the dermis of the skin may potentially reflect the situation within the oral mucosa. Over recent years, the identification of the skin as a local reservoir of adult stem cell populations and the idea that multipotent cell populations exist within the dermal tissues of skin has gained increasing credibility. Indeed, numerous multipotent progenitor cells have been identified within the dermis and resident appendages, all capable of differentiating into multiple cell lineages. Furthermore, a number of these cell populations have been implicated in the repair of these tissues following injury. There is increasing evidence suggesting that such populations of progenitor cells may also reside within the lamina propria. In this respect, the ability to isolate large numbers of multipotent progenitor cells from a tissue which when biopsied heals without a scar would be of great interest scientifically and commercially, particularly with respect to future therapeutic applications and the developing discipline of tissue engineering.
Collapse
Affiliation(s)
- P Stephens
- Wound Biology Group, Cardiff Institute of Tissue Engineering and Repair, Department of Oral Surgery, Medicine & Pathology, School of Dentistry, Cardiff University, Cardiff, UK.
| | | |
Collapse
|