1
|
Bao Y, Tong C, Xiong X. CXCL3: A key player in tumor microenvironment and inflammatory diseases. Life Sci 2024; 348:122691. [PMID: 38714265 DOI: 10.1016/j.lfs.2024.122691] [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: 02/09/2024] [Revised: 04/14/2024] [Accepted: 05/03/2024] [Indexed: 05/09/2024]
Abstract
CXCL3 (C-X-C Motif Chemokine 3), a member of the C-X-C chemokine subfamily, operates as a potent chemoattractant for neutrophils, thereby orchestrating the recruitment and migration of leukocytes alongside eliciting an inflammatory response. Recent inquiries have shed light on the pivotal roles of CXCL3 in the context of carcinogenesis. In the tumor microenvironment, CXCL3 emanating from both tumor and stromal cells intricately modulates cellular behaviors through autocrine and paracrine actions, primarily via interaction with its receptor CXCR2. Activation of signaling cascades such as ERK/MAPK, AKT, and JAK2/STAT3 underscores CXCL3's propensity to favor tumorigenic processes. However, CXCL3 exhibits dualistic behaviors, as evidenced by its capacity to exert anti-tumor effects under specific conditions. Additionally, the involvement of CXCL3 extends to inflammatory disorders like eclampsia, obesity, and asthma. This review encapsulates the structural attributes, biological functionalities, and molecular underpinnings of CXCL3 across both tumorigenesis and inflammatory diseases.
Collapse
Affiliation(s)
- Yuxuan Bao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; Queen Mary School of Nanchang University, Nanchang 330006, China
| | - Chang Tong
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Xiangyang Xiong
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; Province Key Laboratory of Tumor Pathogens and Molecular Pathology, Nanchang University, Nanchang 330006, China.
| |
Collapse
|
2
|
Horwitz A, Birk R. Adipose Tissue Hyperplasia and Hypertrophy in Common and Syndromic Obesity-The Case of BBS Obesity. Nutrients 2023; 15:3445. [PMID: 37571382 PMCID: PMC10421039 DOI: 10.3390/nu15153445] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/16/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Obesity is a metabolic state generated by the expansion of adipose tissue. Adipose tissue expansion depends on the interplay between hyperplasia and hypertrophy, and is mainly regulated by a complex interaction between genetics and excess energy intake. However, the genetic regulation of adipose tissue expansion is yet to be fully understood. Obesity can be divided into common multifactorial/polygenic obesity and monogenic obesity, non-syndromic and syndromic. Several genes related to obesity were found through studies of monogenic non-syndromic obesity models. However, syndromic obesity, characterized by additional features other than obesity, suggesting a more global role of the mutant genes related to the syndrome and, thus, an additional peripheral influence on the development of obesity, were hardly studied to date in this regard. This review summarizes present knowledge regarding the hyperplasia and hypertrophy of adipocytes in common obesity. Additionally, we highlight the scarce research on syndromic obesity as a model for studying adipocyte hyperplasia and hypertrophy, focusing on Bardet-Biedl syndrome (BBS). BBS obesity involves central and peripheral mechanisms, with molecular and mechanistic alternation in adipocyte hyperplasia and hypertrophy. Thus, we argue that using syndromic obesity models, such as BBS, can further advance our knowledge regarding peripheral adipocyte regulation in obesity.
Collapse
Affiliation(s)
| | - Ruth Birk
- Department of Nutrition, Faculty of Health Sciences, Ariel University, Ariel 40700, Israel;
| |
Collapse
|
3
|
van der Sluis N, Scheers EC, Krenning G, van der Lei B, Oonk MH, van Dongen JA. Autologous lipoaspirate as a new treatment of vulvar lichen sclerosus: A review on literature. Exp Dermatol 2022; 31:689-699. [PMID: 35276020 PMCID: PMC9314062 DOI: 10.1111/exd.14561] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/03/2022] [Accepted: 03/05/2022] [Indexed: 11/29/2022]
Abstract
Lichen sclerosus (LS) is a chronic inflammatory dermatosis that mostly affects the genital and anal skin areas. Symptoms may vary from pruritis and pain to sexual dysfunction; however, LS can also be asymptomatic. LS occurs at all ages and in both sexes. Approximately 5% of all women affected by vulvar LS will develop vulvar squamous cell carcinoma. Topical treatment is safe but less effective resulting in chronic course in most patients, who suffer from persistent itching and pain. In severe cases of therapy-resistant LS, there is no adequate treatment. Fat grafting is a novel regenerative therapy to reduce dermal fibrosis. The therapeutic effect of adipose tissue grafts for LS is already investigated in various pioneering studies. This review provides an overview of these studies and the putative mechanisms-of-action of fat grafting to treat LS.
Collapse
Affiliation(s)
- Nanouk van der Sluis
- Department of Plastic SurgeryUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
- Department of Plastic‐, Reconstructive‐ and Hand SurgeryMedisch Spectrum TwenteEnschedeThe Netherlands
| | - Esther C.A.H. Scheers
- Department of Obstetrics and GynecologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Guido Krenning
- Department of Pathology and Medical BiologyUniversity of Groningen and University Medical Center GroningenGroningenThe Netherlands
| | - Berend van der Lei
- Department of Plastic SurgeryUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Maaike H.M. Oonk
- Department of Obstetrics and GynecologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Joris A. van Dongen
- Department of Plastic‐, Reconstructive‐ and Hand Surgery, Utrecht University Medical CenterUtrecht UniversityUtrechtThe Netherlands
| |
Collapse
|
4
|
De la Fuente-Hernandez MA, Alanis-Manriquez EC, Ferat-Osorio E, Rodriguez-Gonzalez A, Arriaga-Pizano L, Vazquez-Santillan K, Melendez-Zajgla J, Fragoso-Ontiveros V, Alvarez-Gomez RM, Maldonado Lagunas V. Molecular changes in adipocyte-derived stem cells during their interplay with cervical cancer cells. Cell Oncol (Dordr) 2022; 45:85-101. [PMID: 35013999 DOI: 10.1007/s13402-021-00653-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2021] [Indexed: 11/03/2022] Open
Abstract
PURPOSE Obesity is as an important risk factor and has been associated with a worse prognosis in at least 13 distinct tumor types. This is partially due to intercellular communication between tumor cells and adipose tissue-derived stem cells (ADSCs), which are increased in obese individuals. As yet, however, little is known about the molecular changes occurring in ADSCs in these conditions. Cervical cancer has a high incidence and mortality rate in women from developing countries, particularly in those with a high body mass index (BMI). METHODS We analyzed the expression profile of ADSCs co-cultured with cervical cancer cells through massive RNA sequencing followed by evaluation of various functional alterations resulting from the modified transcriptome. RESULTS A total of 761 coding and non-coding dysregulated RNAs were identified in ADSCs after co-culture with HeLa cells (validation in CaSki and SiHA cells). Subsequent network analysis showed that these changes were correlated with migration, stemness, DNA repair and cytokine production. Functional experiments revealed a larger ALDHhigh subpopulation and a higher migrative capacity of ADSCs after co-culture with HeLa cells. Interestingly, CXCL3 and its intragenic long-noncoding RNA, lnc-CXCL3, were found to be co-regulated during co-culture. A loss-of-function assay revealed that lnc-CXCL3 acts as a key regulator of CXCL3 expression. CONCLUSIONS Our results suggest that intercellular communication between ADSCs and cervical cancer cells modifies the RNA expression profile in the former, including that of lncRNAs, which in turn can regulate the expression of diverse chemokines that favor malignancy-associated capacities such as migration.
Collapse
Affiliation(s)
- Marcela Angelica De la Fuente-Hernandez
- Facultad de Medicina, Posgrado en Ciencias Biologicas, Universidad Nacional Autonoma de Mexico (UNAM), Av. Ciudad Universitaria 3000, C.P. 04510, Coyoacan, Mexico City, Mexico.,Epigenetics Laboratory, Instituto Nacional de Medicina Genomica (INMEGEN), Periferico Sur No. 4809, Col. Arenal Tepepan, Tlalpan, C.P, 14610, Mexico City, Mexico
| | - Erika Claudia Alanis-Manriquez
- Epigenetics Laboratory, Instituto Nacional de Medicina Genomica (INMEGEN), Periferico Sur No. 4809, Col. Arenal Tepepan, Tlalpan, C.P, 14610, Mexico City, Mexico
| | - Eduardo Ferat-Osorio
- Gastrosurgery Service, UMAE. Hospital de Especialidades Dr. Bernardo Sepulveda Gutierrez of the Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Av. Cuauhtemoc No 330, Col. Doctores, Cuauhtemoc, C.P., 06720, Mexico City, Mexico
| | - Arturo Rodriguez-Gonzalez
- Gastrosurgery Service, UMAE. Hospital de Especialidades Dr. Bernardo Sepulveda Gutierrez of the Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Av. Cuauhtemoc No 330, Col. Doctores, Cuauhtemoc, C.P., 06720, Mexico City, Mexico
| | - Lourdes Arriaga-Pizano
- Unidad de Investigacion Medica en Inmunoquimica. Hospital de Especialidades, Dr. Bernardo Sepulveda Gutierrez of the Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Av. Cuauhtemoc No 330, Col. Doctores, Cuauhtemoc, C.P., 06720, Mexico City, Mexico
| | - Karla Vazquez-Santillan
- Epigenetics Laboratory, Instituto Nacional de Medicina Genomica (INMEGEN), Periferico Sur No. 4809, Col. Arenal Tepepan, Tlalpan, C.P, 14610, Mexico City, Mexico
| | - Jorge Melendez-Zajgla
- Functional Cancer Genomics Laboratory, Instituto Nacional de Medicina Genomica (INMEGEN), Periférico Sur No. 4809, Col. Arenal Tepepan, Tlalpan, C.P., 14610, Mexico City, Mexico
| | | | | | - Vilma Maldonado Lagunas
- Facultad de Medicina, Posgrado en Ciencias Biologicas, Universidad Nacional Autonoma de Mexico (UNAM), Av. Ciudad Universitaria 3000, C.P. 04510, Coyoacan, Mexico City, Mexico. .,Epigenetics Laboratory, Instituto Nacional de Medicina Genomica (INMEGEN), Periferico Sur No. 4809, Col. Arenal Tepepan, Tlalpan, C.P, 14610, Mexico City, Mexico.
| |
Collapse
|
5
|
Bou Malhab LJ, Abdel-Rahman WM. Obesity and inflammation: colorectal cancer engines. Curr Mol Pharmacol 2021; 15:620-646. [PMID: 34488607 DOI: 10.2174/1874467214666210906122054] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 05/14/2021] [Accepted: 05/17/2021] [Indexed: 12/24/2022]
Abstract
The prevalence of obesity continues to increase to the extent that it became a worldwide pandemic. An accumulating body of evidence has associated obesity with the development of different types of cancer, including colorectal cancer, which is a notorious disease with a high mortality rate. At the molecular level, colorectal cancer is a heterogenous disease characterized by a myriad of genetic and epigenetic alterations associated with various forms of genomic instability (detailed in Supplementary Materials). Recently, the microenvironment has emerged as a major factor in carcinogenesis. Our aim is to define the different molecular alterations leading to the development of colorectal cancer in obese patients with a focus on the role of the microenvironment in carcinogenesis. We also highlight all existent molecules in clinical trials that target the activated pathways in obesity-associated colorectal cancer, whether used as single treatments or in combination. Obesity predisposes to colorectal cancer via creating a state of chronic inflammation with dysregulated adipokines, inflammatory mediators, and other factors such as immune cell infiltration. A unifying theme in obesity-mediated colorectal cancer is the activation of the PI3K/AKT, mTOR/MAPK, and STAT3 signaling pathways. Different inhibitory molecules towards these pathways exist, increasing the therapeutic choice of obesity-associated colon cancer. However, obese patients are more likely to suffer from chemotherapy overdosing. Preventing obesity through maintaining a healthy and active lifestyle remains to be the best remedy.
Collapse
Affiliation(s)
- Lara J Bou Malhab
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah. United Arab Emirates
| | - Wael M Abdel-Rahman
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah. United Arab Emirates
| |
Collapse
|
6
|
Iranparast S, Tayebi S, Ahmadpour F, Yousefi B. Tumor-Induced Metabolism and T Cells Located in Tumor Environment. Curr Cancer Drug Targets 2020; 20:741-756. [PMID: 32691710 DOI: 10.2174/1568009620666200720010647] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/30/2020] [Accepted: 05/04/2020] [Indexed: 12/14/2022]
Abstract
Several subtypes of T cells are located in a tumor environment, each of which supplies their energy using different metabolic mechanisms. Since the cancer cells require high levels of glucose, the conditions of food poverty in the tumor environment can cause inactivation of immune cells, especially the T-effector cells, due to the need for glucose in the early stages of these cells activity. Different signaling pathways, such as PI3K-AKt-mTOR, MAPK, HIF-1α, etc., are activated or inactivated by the amount and type of energy source or oxygen levels that determine the fate of T cells in a cancerous environment. This review describes the metabolites in the tumor environment and their effects on the function of T cells. It also explains the signaling pathway of T cells in the tumor and normal conditions, due to the level of access to available metabolites and subtypes of T cells in the tumor environment.
Collapse
Affiliation(s)
- Sara Iranparast
- Department of Immunology, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran,Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sanaz Tayebi
- Department of Immunology, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Fatemeh Ahmadpour
- Department of Biochemistry, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
7
|
PAI-1-Dependent Inactivation of SMAD4-Modulated Junction and Adhesion Complex in Obese Endometrial Cancer. Cell Rep 2020; 33:108253. [PMID: 33053339 PMCID: PMC7641039 DOI: 10.1016/j.celrep.2020.108253] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 07/01/2020] [Accepted: 09/17/2020] [Indexed: 12/03/2022] Open
Abstract
While plasminogen activator inhibitor-1 (PAI-1) is known to potentiate cellular migration via proteolytic regulation, this adipokine is implicated as an oncogenic ligand in the tumor microenvironment. To understand the underlying paracrine mechanism, here, we conduct transcriptomic analysis of 1,898 endometrial epithelial cells (EECs) exposed and unexposed to PAI-1-secreting adipose stromal cells. The PAI-1-dependent action deregulates crosstalk among tumor-promoting and tumor-repressing pathways, including transforming growth factor β (TGF-β). When PAI-1 is tethered to lipoprotein receptor-related protein 1 (LRP1), the internalized signaling causes downregulation of SMAD4 at the transcriptional and post-translational levels that attenuates TGF-β-related transcription programs. Repression of genes encoding the junction and adhesion complex preferentially occurs in SMAD4-underexpressed EECs of persons with obesity. The findings highlight a role of PAI-1 signaling that renders ineffective intercellular communication for the development of adiposity-associated endometrial cancer. Lin et al. demonstrate that PAI-1 secreted by adipose stromal cells interacts with LRP1 to repress TGF-β/SMAD4-regulated genes linked to cellular junction and adhesion complexes. This action disrupts cell-cell communication and facilitates the development of obesity-driven endometrial cancer.
Collapse
|
8
|
Teufelsbauer M, Rath B, Plangger A, Staud C, Nanobashvili J, Huk I, Neumayer C, Hamilton G, Radtke C. Effects of metformin on adipose-derived stromal cell (ADSC) - Breast cancer cell lines interaction. Life Sci 2020; 261:118371. [PMID: 32882267 DOI: 10.1016/j.lfs.2020.118371] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 08/28/2020] [Accepted: 08/28/2020] [Indexed: 10/23/2022]
Abstract
AIMS Metformin is a clinical drug administered to patients to treat type 2 diabetes mellitus that was found to be associated with a lower risk of occurrence of cancer and cancer-related death. The present study investigated the effects of metformin on human adipose-derived stromal cells (ADSC) - breast cancer cell line interactions. MAIN METHODS ADSCs grown from lipoaspirates were tested for growth-stimulating and migration-controlling activity on breast cancer cell lines after pretreatment with metformin. Furthermore, secreted proteins of ADSCs, phosphorylation of intracellular proteins and the effect of metformin on adipocytic differentiation of ADSCs were assayed. KEY FINDINGS Compared to breast cancer cell lines (4.0 ± 3.5% reduction of proliferation), 2 mM metformin significantly inhibited the proliferation of ADSC lines (19.2 ± 8.4% reduction of proliferation). This effect on ADSCs seems to be mediated by altered phosphorylation of GSK-3, CREB and PRAS40. Furthermore, treatment with metformin abolished the induction of differentiation of three ADSC lines to adipocytes. 1 and 2 mM metformin significantly impaired the migration of breast cancer cell lines MDA-MB-231 and MDA-MB-436 in scratch assays. SIGNIFICANCE Metformin showed low direct inhibitory effects on breast cancer cell lines at physiological concentrations but exerted a significant retardation of the growth and the adipocytic differentiation of ADSCs. Thus, the anticancer activity of metformin in breast cancer at physiological drug concentrations seems to be mediated by an indirect mechanism that lowers the supportive activity of ADSCs.
Collapse
Affiliation(s)
- Maryana Teufelsbauer
- Department of Plastic and Reconstructive Surgery, Medical University of Vienna, Vienna, Austria
| | - Barbara Rath
- Department of Vascular Surgery, Medical University of Vienna, Vienna, Austria
| | - Adelina Plangger
- Department of Vascular Surgery, Medical University of Vienna, Vienna, Austria
| | - Clement Staud
- Department of Plastic and Reconstructive Surgery, Medical University of Vienna, Vienna, Austria
| | - Josif Nanobashvili
- Department of Vascular Surgery, Medical University of Vienna, Vienna, Austria
| | - Ihor Huk
- Department of Vascular Surgery, Medical University of Vienna, Vienna, Austria
| | - Christoph Neumayer
- Department of Vascular Surgery, Medical University of Vienna, Vienna, Austria
| | - Gerhard Hamilton
- Department of Vascular Surgery, Medical University of Vienna, Vienna, Austria.
| | - Christine Radtke
- Department of Plastic and Reconstructive Surgery, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
9
|
Wang T, Yu X, Lin J, Qin C, Bai T, Xu T, Wang L, Liu X, Li S. Adipose-Derived Stem Cells Inhibited the Proliferation of Bladder Tumor Cells by S Phase Arrest and Wnt/β-Catenin Pathway. Cell Reprogram 2020; 21:331-338. [PMID: 31809208 PMCID: PMC6918853 DOI: 10.1089/cell.2019.0047] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Adipose-derived stem cells (ADSCs), which are present in most organs and tissues, were evaluated as a novel medium for stem cell therapy. In this study, we investigated the effects and underlying mechanisms of ADSCs in bladder tumor (BT) cells. SV-HUC, T24, and EJ cells were cultured with ADSCs and conditioned medium from ADSCs (ADSC-CM). We observed that in routine culture, ADSCs significantly inhibited the proliferation of T24 and EJ cells in a dose-dependent manner. In addition, ADSC-CM attenuated the viability of T24 and EJ cells in a dose-dependent manner. Cell cycle analysis indicated that ADSC-CM was capable of inducing T24 and EJ cells S phase arrest and downregulating the expression of CDK 1, whereas the expression of cyclin A was increased. ADSC-CM could induce apoptosis in T24 cells. The mechanism of this effect likely involved the caspase3/7 pathway and Wnt/β-catenin pathway. These findings demonstrated that ADSCs could inhibit the proliferation of BT cells via secretory factors.
Collapse
Affiliation(s)
- Tao Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xi Yu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jian Lin
- Department of Urology, Peking University First Hospital, Beijing, China
| | - Cong Qin
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tao Bai
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tao Xu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lei Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiuheng Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shenglan Li
- Department of Radiography, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
10
|
Eckel-Mahan K, Ribas Latre A, Kolonin MG. Adipose Stromal Cell Expansion and Exhaustion: Mechanisms and Consequences. Cells 2020; 9:cells9040863. [PMID: 32252348 PMCID: PMC7226766 DOI: 10.3390/cells9040863] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 03/12/2020] [Accepted: 03/17/2020] [Indexed: 12/13/2022] Open
Abstract
Adipose tissue (AT) is comprised of a diverse number of cell types, including adipocytes, stromal cells, endothelial cells, and infiltrating leukocytes. Adipose stromal cells (ASCs) are a mixed population containing adipose progenitor cells (APCs) as well as fibro-inflammatory precursors and cells supporting the vasculature. There is growing evidence that the ability of ASCs to renew and undergo adipogenesis into new, healthy adipocytes is a hallmark of healthy fat, preventing disease-inducing adipocyte hypertrophy and the spillover of lipids into other organs, such as the liver and muscles. However, there is building evidence indicating that the ability for ASCs to self-renew is not infinite. With rates of ASC proliferation and adipogenesis tightly controlled by diet and the circadian clock, the capacity to maintain healthy AT via the generation of new, healthy adipocytes appears to be tightly regulated. Here, we review the contributions of ASCs to the maintenance of distinct adipocyte pools as well as pathogenic fibroblasts in cancer and fibrosis. We also discuss aging and diet-induced obesity as factors that might lead to ASC senescence, and the consequences for metabolic health.
Collapse
Affiliation(s)
- Kristin Eckel-Mahan
- Institute of Molecular Medicine, McGovern Medical School at the University of Texas Health Science Center, Houston, TX 77030, USA;
| | - Aleix Ribas Latre
- Helmholtz Institute for Metabolic, Obesity and Vascular Research Center, D-04103 Leipzig, Germany;
| | - Mikhail G. Kolonin
- Institute of Molecular Medicine, McGovern Medical School at the University of Texas Health Science Center, Houston, TX 77030, USA;
- Correspondence:
| |
Collapse
|
11
|
Gentile P, Calabrese C, De Angelis B, Pizzicannella J, Kothari A, Garcovich S. Impact of the Different Preparation Methods to Obtain Human Adipose-Derived Stromal Vascular Fraction Cells (AD-SVFs) and Human Adipose-Derived Mesenchymal Stem Cells (AD-MSCs): Enzymatic Digestion Versus Mechanical Centrifugation. Int J Mol Sci 2019; 20:E5471. [PMID: 31684107 PMCID: PMC6862236 DOI: 10.3390/ijms20215471] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 10/27/2019] [Accepted: 11/01/2019] [Indexed: 12/16/2022] Open
Abstract
Autologous therapies using adipose-derived stromal vascular fraction (AD-SVFs) and adult adipose-derived mesenchymal stem cells (AD-MSCs) warrant careful preparation of the harvested adipose tissue. Currently, no standardized technique for this preparation exists. Processing quantitative standards (PQSs) define manufacturing quantitative variables (such as time, volume, and pressure). Processing qualitative standards (PQLSs) define the quality of the materials and methods in manufacturing. The purpose of the review was to use PQSs and PQLSs to report the in vivo and in vitro results obtained by different processing kits that use different procedures (enzymatic vs. non-enzymatic) to isolate human AD-SVFs/AD-MSCs. PQSs included the volume of fat tissue harvested and reagents used, the time/gravity of centrifugation, and the time, temperature, and tilt level/speed of incubation and/or centrifugation. PQLSs included the use of a collagenase, a processing time of 30 min, kit weight, transparency of the kit components, the maintenance of a closed sterile processing environment, and the use of a small centrifuge and incubating rocker. Using a kit with the PQSs and PQLSs described in this study enables the isolation of AD-MSCs that meet the consensus quality criteria. As the discovery of new critical quality attributes (CQAs) of AD-MSCs evolve with respect to purity and potency, adjustments to these benchmark PQSs and PQLs will hopefully isolate AD-MSCs of various CQAs with greater reproducibility, quality, and safety. Confirmatory studies will no doubt need to be completed.
Collapse
Affiliation(s)
- Pietro Gentile
- Surgical Science Department, Plastic and Reconstructive Surgery, University of Rome "Tor Vergata", 00179 Rome, Italy.
| | | | - Barbara De Angelis
- Surgical Science Department, Plastic and Reconstructive Surgery, University of Rome "Tor Vergata", 00179 Rome, Italy.
| | | | - Ashutosh Kothari
- Chief of Breast Surgery Unit, Guy's Hospital, Guy's and St. Thomas' NHS Foundation Trust, London SE1 9RT, UK.
| | - Simone Garcovich
- Institute of Dermatology, F. Policlinico Gemelli IRCSS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| |
Collapse
|
12
|
Mohanty SS, Mohanty PK. Obesity as potential breast cancer risk factor for postmenopausal women. Genes Dis 2019; 8:117-123. [PMID: 33997158 PMCID: PMC8099684 DOI: 10.1016/j.gendis.2019.09.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 08/21/2019] [Accepted: 09/04/2019] [Indexed: 12/15/2022] Open
Abstract
Breast cancer is the second highest prevalent cancer globally after lung cancer with 2.09 million cases during 2018. Adults about 1.9 billion were overweight and over 650 million out of these were obese during 2016. There is a significant relationship between breast cancer risk and obesity. Premature menopause and premenopausal obesity diminish the risk whereas postmenopausal obesity amplifies the risk, because adipose tissue acts as the major reservoir for estrogen biosynthesis after menopause. Lofty estrogen levels in serum along with enhanced peripheral site production of estrogen have been viewed as major reasons of developing breast cancer in overweight postmenopausal women. This review explains body fat as a peripheral site for estrogen biosynthesis, estrogen exposure affecting body fat distribution, and the mechanism of estrogen production from body fats.
Collapse
Affiliation(s)
- Swati Sucharita Mohanty
- Cytogenetics Laboratory, P.G. Department of Zoology, Utkal University, Bhubaneswar, 751004, Odisha, India
| | - Prafulla Kumar Mohanty
- Cytogenetics Laboratory, P.G. Department of Zoology, Utkal University, Bhubaneswar, 751004, Odisha, India
| |
Collapse
|
13
|
Scioli MG, Storti G, D'Amico F, Gentile P, Kim BS, Cervelli V, Orlandi A. Adipose-Derived Stem Cells in Cancer Progression: New Perspectives and Opportunities. Int J Mol Sci 2019; 20:ijms20133296. [PMID: 31277510 PMCID: PMC6651808 DOI: 10.3390/ijms20133296] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/01/2019] [Accepted: 07/02/2019] [Indexed: 12/12/2022] Open
Abstract
Growing importance has been attributed to interactions between tumors, the stromal microenvironment and adult mesenchymal stem cells. Adipose-derived stem cells (ASCs) are routinely employed in regenerative medicine and in autologous fat transfer procedures. To date, clinical trials have failed to demonstrate the potential pro-oncogenic role of ASC enrichment. Nevertheless, some pre-clinical studies from in vitro and in vivo models have suggested that ASCs act as a potential tumor promoter for different cancer cell types, and support tumor progression and invasiveness through the activation of several intracellular signals. Interaction with the tumor microenvironment and extracellular matrix remodeling, the exosomal release of pro-oncogenic factors as well as the induction of epithelial-mesenchymal transitions are the most investigated mechanisms. Moreover, ASCs have also demonstrated an elective tumor homing capacity and this tumor-targeting capacity makes them a suitable carrier for anti-cancer drug delivery. New genetic and applied nanotechnologies may help to design promising anti-cancer cell-based approaches through the release of loaded intracellular nanoparticles. These new anti-cancer therapies can more effectively target tumor cells, reaching higher local concentrations even in pharmacological sanctuaries, and thus minimizing systemic adverse drug effects. The potential interplay between ASCs and tumors and potential ASCs-based therapeutic approaches are discussed.
Collapse
Affiliation(s)
- Maria Giovanna Scioli
- Anatomic Pathology Institute, Department of Biomedicine and Prevention, Tor Vergata University of Rome, 00133 Rome, Italy
| | - Gabriele Storti
- Plastic and Reconstructive Surgery, Department of Surgical Sciences, Tor Vergata University of Rome, 00133 Rome, Italy
| | - Federico D'Amico
- Anatomic Pathology Institute, Department of Biomedicine and Prevention, Tor Vergata University of Rome, 00133 Rome, Italy
| | - Pietro Gentile
- Plastic and Reconstructive Surgery, Department of Surgical Sciences, Tor Vergata University of Rome, 00133 Rome, Italy
| | - Bong-Sung Kim
- Division of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Valerio Cervelli
- Plastic and Reconstructive Surgery, Department of Surgical Sciences, Tor Vergata University of Rome, 00133 Rome, Italy
| | - Augusto Orlandi
- Anatomic Pathology Institute, Department of Biomedicine and Prevention, Tor Vergata University of Rome, 00133 Rome, Italy.
| |
Collapse
|
14
|
Gentile P, Garcovich S. Concise Review: Adipose-Derived Stem Cells (ASCs) and Adipocyte-Secreted Exosomal microRNA (A-SE-miR) Modulate Cancer Growth and proMote Wound Repair. J Clin Med 2019; 8:jcm8060855. [PMID: 31208047 PMCID: PMC6616456 DOI: 10.3390/jcm8060855] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 06/05/2019] [Accepted: 06/12/2019] [Indexed: 12/13/2022] Open
Abstract
Adipose-derived stem cells (ASCs) have been routinely used from several years in regenerative surgery without any definitive statement about their potential pro-oncogenic or anti-oncogenic role. ASCs has proven to favor tumor progression in several experimental cancer models, playing a central role in regulating tumor invasiveness and metastatic potential through several mechanisms, such as the paracrine release of exosomes containing pro-oncogenic molecules and the induction of epithelial-mesenchymal transition. However, the high secretory activity and the preferential tumor-targeting make also ASCs a potentially suitable vehicle for delivery of new anti-cancer molecules in tumor microenvironment. Nanotechnologies, viral vectors, drug-loaded exosomes, and micro-RNAs (MiR) represent additional new tools that can be applied for cell-mediated drug delivery in a tumor microenvironment. Recent studies revealed that the MiR play important roles in paracrine actions on adipose-resident macrophages, and their dysregulation has been implicated in the pathogenesis of obesity, diabetes, and diabetic complications as wounds. Numerous MiR are present in adipose tissues, actively participating in the regulation of adipogenesis, adipokine secretion, inflammation, and inter-cellular communications in the local tissues. These results provide important insights into Adipocyte-secreted exosomal microRNA (A-SE-MiR) function and they suggest evaluating the potential role of A-SE-MiR in tumor progression, the mechanisms underlying ASCs-cancer cell interplay and clinical safety of ASCs-based therapies.
Collapse
Affiliation(s)
- Pietro Gentile
- Surgical Science Department, Plastic and Reconstructive Surgery Unit, University of "Tor Vergata", 00133 Rome, Italy.
| | - Simone Garcovich
- Institute of Dermatology, F. Policlinico Gemelli IRCSS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| |
Collapse
|
15
|
Di Stefano AB, Massihnia D, Grisafi F, Castiglia M, Toia F, Montesano L, Russo A, Moschella F, Cordova A. Adipose tissue, angiogenesis and angio-MIR under physiological and pathological conditions. Eur J Cell Biol 2019; 98:53-64. [DOI: 10.1016/j.ejcb.2018.11.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 11/28/2018] [Accepted: 11/29/2018] [Indexed: 01/06/2023] Open
|
16
|
Polusani SR, Huang YW, Huang G, Chen CW, Wang CM, Lin LL, Osmulski P, Lucio ND, Liu L, Hsu YT, Zhou Y, Lin CL, Aguilera-Barrantes I, Valente PT, Kost ER, Chen CL, Shim EY, Lee SE, Ruan J, Gaczynska ME, Yan P, Goodfellow PJ, Mutch DG, Jin VX, Nicholson BJ, Huang THM, Kirma NB. Adipokines Deregulate Cellular Communication via Epigenetic Repression of Gap Junction Loci in Obese Endometrial Cancer. Cancer Res 2019; 79:196-208. [PMID: 30389702 PMCID: PMC6705596 DOI: 10.1158/0008-5472.can-18-1615] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 09/10/2018] [Accepted: 10/26/2018] [Indexed: 11/16/2022]
Abstract
Emerging evidence indicates that adipose stromal cells (ASC) are recruited to enhance cancer development. In this study, we examined the role these adipocyte progenitors play relating to intercellular communication in obesity-associated endometrial cancer. This is particularly relevant given that gap junctions have been implicated in tumor suppression. Examining the effects of ASCs on the transcriptome of endometrial epithelial cells (EEC) in an in vitro coculture system revealed transcriptional repression of GJA1 (encoding the gap junction protein Cx43) and other genes related to intercellular communication. This repression was recapitulated in an obesity mouse model of endometrial cancer. Furthermore, inhibition of plasminogen activator inhibitor 1 (PAI-1), which was the most abundant ASC adipokine, led to reversal of cellular distribution associated with the GJA1 repression profile, suggesting that PAI-1 may mediate actions of ASC on transcriptional regulation in EEC. In an endometrial cancer cohort (n = 141), DNA hypermethylation of GJA1 and related loci TJP2 and PRKCA was observed in primary endometrial endometrioid tumors and was associated with obesity. Pharmacologic reversal of DNA methylation enhanced gap-junction intercellular communication and cell-cell interactions in vitro. Restoring Cx43 expression in endometrial cancer cells reduced cellular migration; conversely, depletion of Cx43 increased cell migration in immortalized normal EEC. Our data suggest that persistent repression by ASC adipokines leads to promoter hypermethylation of GJA1 and related genes in the endometrium, triggering long-term silencing of these loci in endometrial tumors of obese patients. SIGNIFICANCE: Studies reveal that adipose-derived stem cells in endometrial cancer pathogenesis influence epigenetic repression of gap junction loci, which suggests targeting of gap junction activity as a preventive strategy for obesity-associated endometrial cancer.
Collapse
Affiliation(s)
- Srikanth R Polusani
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, Texas
| | - Yi-Wen Huang
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukie, Wisconsin
| | - Guangcun Huang
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, Texas
| | - Chun-Wei Chen
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, Texas
| | - Chiou-Miin Wang
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, Texas
| | - Li-Ling Lin
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, Texas
| | - Pawel Osmulski
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, Texas
| | - Nicholas D Lucio
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, Texas
| | - Lu Liu
- Department of Computer Science, North Dakota State University, Fargo, North Dakota
| | - Ya-Ting Hsu
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, Texas
| | - Yufan Zhou
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, Texas
| | - Chun-Lin Lin
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, Texas
| | | | - Philip T Valente
- Department of Pathology, University of Texas Health San Antonio, San Antonio, Texas
| | - Edward R Kost
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, Texas
| | - Chun-Liang Chen
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, Texas
| | - Eun Yong Shim
- Department of Radiation Oncology, University of Texas Health San Antonio, San Antonio, Texas
| | - Sang Eun Lee
- Department of Radiation Oncology, University of Texas Health San Antonio, San Antonio, Texas
| | - Jianhua Ruan
- Department of Computer Science, University of Texas San Antonio, San Antonio, Texas
| | - Maria E Gaczynska
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, Texas
| | - Pearlly Yan
- Department of Internal Medicine, Ohio State University, Columbus, Ohio
| | - Paul J Goodfellow
- Department of Obstetrics and Gynecology, Ohio State University, Columbus, Ohio
| | - David G Mutch
- Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri
| | - Victor X Jin
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, Texas
| | - Bruce J Nicholson
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas
| | - Tim H-M Huang
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, Texas.
| | - Nameer B Kirma
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, Texas.
| |
Collapse
|
17
|
Obesity and gastrointestinal cancer: the interrelationship of adipose and tumour microenvironments. Nat Rev Gastroenterol Hepatol 2018; 15:699-714. [PMID: 30323319 DOI: 10.1038/s41575-018-0069-7] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Increasing recognition of an association between obesity and many cancer types exists, but how the myriad of local and systemic effects of obesity affect key cellular and non-cellular processes within the tumour microenvironment (TME) relevant to carcinogenesis, tumour progression and response to therapies remains poorly understood. The TME is a complex cellular environment in which the tumour exists along with blood vessels, immune cells, fibroblasts, bone marrow-derived inflammatory cells, signalling molecules and the extracellular matrix. Obesity, in particular visceral obesity, might fuel the dysregulation of key pathways relevant to both the adipose microenvironment and the TME, which interact to promote carcinogenesis in at-risk epithelium. The tumour-promoting effects of obesity can occur at the local level as well as systemically via circulating inflammatory, growth factor and metabolic mediators associated with adipose tissue inflammation, as well as paracrine and autocrine effects. This Review explores key pathways linking visceral obesity and gastrointestinal cancer, including inflammation, hypoxia, altered stromal and immune cell function, energy metabolism and angiogenesis.
Collapse
|
18
|
Recent Advances and Future Directions in Postmastectomy Breast Reconstruction. Clin Breast Cancer 2018; 18:e571-e585. [DOI: 10.1016/j.clbc.2018.02.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 02/08/2018] [Accepted: 02/10/2018] [Indexed: 11/20/2022]
|
19
|
Gérard C, Brown KA. Obesity and breast cancer - Role of estrogens and the molecular underpinnings of aromatase regulation in breast adipose tissue. Mol Cell Endocrinol 2018; 466:15-30. [PMID: 28919302 DOI: 10.1016/j.mce.2017.09.014] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Revised: 09/12/2017] [Accepted: 09/13/2017] [Indexed: 12/15/2022]
Abstract
One in eight women will develop breast cancer over their lifetime making it the most common female cancer. The cause of breast cancer is multifactorial and includes hormonal, genetic and environmental cues. Obesity is now an accepted risk factor for breast cancer in postmenopausal women, particularly for the hormone-dependent subtype of breast cancer. Obesity, which is characterized by an excess accumulation of body fat, is at the origin of chronic inflammation of white adipose tissue and is associated with dramatic changes in the biology of adipocytes leading to their dysfunction. Inflammatory factors found in the breast of obese women considerably impact estrogen signaling, mainly by driving changes in aromatase expression the enzyme responsible for estrogen production, and therefore promote tumor formation and progression. There is thus a strong link between adipose inflammation and estrogen biosynthesis and their signaling pathways converge in obese patients. This review describes how obesity-related factors can affect the risk of hormone-dependent breast cancer, highlighting the different molecular mechanisms and metabolic pathways involved in aromatase regulation, estrogen production and breast malignancy in the context of obesity.
Collapse
Affiliation(s)
- Céline Gérard
- Metabolism & Cancer Laboratory, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Kristy A Brown
- Metabolism & Cancer Laboratory, Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Physiology, Monash University, Clayton, VIC, Australia; Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
20
|
Cao H, Huang Y, Wang L, Wang H, Pang X, Li K, Dang W, Tang H, Wei L, Su M, Tang C, Chen T. Leptin promotes migration and invasion of breast cancer cells by stimulating IL-8 production in M2 macrophages. Oncotarget 2018; 7:65441-65453. [PMID: 27588409 PMCID: PMC5323167 DOI: 10.18632/oncotarget.11761] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Accepted: 08/13/2016] [Indexed: 02/02/2023] Open
Abstract
This study aims to investigate the mechanisms underlying leptin-mediated crosstalk between tumor-associated macrophages (M2 macrophages) and breast cancer cells. THP1 human leukemic monocytes were induced to differentiate into M2 macrophages by PMA (100 nM) and IL-4 (20 ng/mL). Quantitative RT-PCR and Western blot revealed that leptin (100 nM) significantly increased the expression of leptin receptor (ObR) in the M2 macrophages (P < 0.01) and stimulated interleukin (IL)-8 expression in the M2 macrophages, mouse macrophage cells RAW264.7, and primary mouse peritoneal macrophages in a dose- and time-dependent manner. Leptin-induced IL-8 production was sensitive to the ERK inhibitor PD980590 (10 μmol/L), p38 MAPK inhibitor SB203580 (20 μmol/L), and anti-ObR neutralizing antibody (4 μg/mL). Leptin (100 ng/mL) substantially increased the phosphorylation of p38 and ERK1/2. Thus, leptin may induce IL-8 production in M2 macrophages by interacting with ObR to activate the p38 and ERK signaling pathways. Scratch and transwell chamber assay showed that both recombinant IL-8 and leptin-induced M2 macrophage-derived IL-8 promoted the migration and invasion of human breast cancer cells MCF7 and MDA-MB-231 (All P < 0.01). In a nude mice xenograft model of breast cancer (n = 5 per group), injection of leptin (0.1 μg/g) dramatically increased tumor volume and mass, reduced survival, exacerbated pulmonary metastasis, and elevated IL-8 and Ki67 expression in the tumor tissue (All P < 0.05) compared with PBS injection. Depletion of mouse macrophage by Clophosome®-clodronate liposome and injection of anti-mouse IL-8 neutralizing antibodies in the xenograft tumor significantly attenuated those leptin-mediated stimulations (All P < 0.05). These findings indicate that leptin may promote tumor growth and metastasis by stimulating IL-8 production in tumor-associated macrophage.
Collapse
Affiliation(s)
- Hong Cao
- Department of Laboratory Medicine Key Laboratory of Diagnostic Medicine, Ministry of Education Chongqing Medical University, Chongqing, China
| | - Yunxiu Huang
- Department of Laboratory Medicine Key Laboratory of Diagnostic Medicine, Ministry of Education Chongqing Medical University, Chongqing, China
| | - Lin Wang
- Department of Laboratory Medicine Key Laboratory of Diagnostic Medicine, Ministry of Education Chongqing Medical University, Chongqing, China
| | - Hong Wang
- Department of Laboratory Medicine Key Laboratory of Diagnostic Medicine, Ministry of Education Chongqing Medical University, Chongqing, China
| | - Xueli Pang
- Department of Laboratory Medicine Key Laboratory of Diagnostic Medicine, Ministry of Education Chongqing Medical University, Chongqing, China
| | - Kuangfa Li
- Department of Laboratory Medicine Key Laboratory of Diagnostic Medicine, Ministry of Education Chongqing Medical University, Chongqing, China
| | - Weiqi Dang
- Department of Laboratory Medicine Key Laboratory of Diagnostic Medicine, Ministry of Education Chongqing Medical University, Chongqing, China
| | - Hao Tang
- Department of Laboratory Medicine Key Laboratory of Diagnostic Medicine, Ministry of Education Chongqing Medical University, Chongqing, China
| | - Lan Wei
- Department of Laboratory Medicine Key Laboratory of Diagnostic Medicine, Ministry of Education Chongqing Medical University, Chongqing, China
| | - Min Su
- Department of Laboratory Medicine Key Laboratory of Diagnostic Medicine, Ministry of Education Chongqing Medical University, Chongqing, China
| | - Cuiping Tang
- Department of Laboratory Medicine Key Laboratory of Diagnostic Medicine, Ministry of Education Chongqing Medical University, Chongqing, China
| | - Tingmei Chen
- Department of Laboratory Medicine Key Laboratory of Diagnostic Medicine, Ministry of Education Chongqing Medical University, Chongqing, China
| |
Collapse
|
21
|
Carraro A, Trevellin E, Fassan M, Kotsafti A, Lunardi F, Porzionato A, Dall'Olmo L, Cagol M, Alfieri R, Macchi V, Tedeschi U, Calabrese F, Rugge M, Castoro C, Vettor R, Scarpa M. Esophageal adenocarcinoma microenvironment: Peritumoral adipose tissue effects associated with chemoresistance. Cancer Sci 2017; 108:2393-2404. [PMID: 28985034 PMCID: PMC5715298 DOI: 10.1111/cas.13415] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 09/29/2017] [Accepted: 10/02/2017] [Indexed: 12/17/2022] Open
Abstract
Peritumoral microenvironment affects cancer development and chemoresistance, and visceral adipose tissue may play a critical role. We aimed to identify depot‐specific adipose characteristics associated with carcinogenesis and resistance to neoadjuvant therapy in esophageal adenocarcinoma (EAC). We analyzed: (i) the peritumoral adipose tissue of rats following the induction of esophageal carcinogenesis; (ii) the peritumoral and distal (omental) adipose tissue of patients affected by EAC; (iii) adipose‐derived stem cells (ADSC) isolated from healthy patients and treated with conditioned medium (CM), collected from tumoral and adipose tissue of patients with EAC. In peritumoral adipose tissue of rats, CD34, CD31 and vascular endothelial growth factor (VEGF) expression increased progressively during EAC development. In patients with EAC, expression of CD34, CD45, CD90 and nucleostemin (NSTM) was higher in peritumoral than in distal adipose tissue and decreased in the presence of neoadjuvant therapy. Moreover, expression of NSTM, octamer‐binding transcription factor 4 (OCT‐4) and VEGF was higher in peritumoral (but not in distal) adipose tissue of chemoresistant patients. In ADSC, treatment with peritumoral adipose tissue CM increased the adipogenic potential and the expression of CD34, CD90, NSTM and OCT‐4. These effects were similar to those induced by cancer‐derived CM, but were not observed in ADSC treated with distal adipose tissue CM and were partially reduced by a leptin antagonist. Last, ADSC treated with peritumoral CM of chemoresistant patients displayed increased expression of NSTM, OCT‐4, leptin, leptin receptor, alpha‐smooth muscle actin (α‐SMA), CD34 and VEGF. These results suggest that peritumoral adipose tissue may promote, by paracrine signaling, the expression of depot‐specific factors associated with therapeutic resistance.
Collapse
Affiliation(s)
- Amedeo Carraro
- Department of General Surgery and Odontoiatrics, University Hospital of Verona, Verona, Italy
| | - Elisabetta Trevellin
- Department of Medicine, Endocrine-Metabolic Laboratory, University of Padova, Padova, Italy
| | - Matteo Fassan
- Department of Medicine, Surgical Pathology & Cytopathology Unit, University of Padova, Padova, Italy
| | - Andromachi Kotsafti
- Esophageal and Digestive Tract Surgical Unit, Veneto Institute of Oncology (IOV-IRCCS), Padova, Italy
| | - Francesca Lunardi
- Department of Cardiothoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Andrea Porzionato
- Department of Molecular Medicine, Normal Anatomy Unit, University of Padova, Padova, Italy
| | - Luigi Dall'Olmo
- Department of Emergency Medicine, "Santi Giovanni e Paolo" Hospital, Venice, Italy
| | - Matteo Cagol
- Esophageal and Digestive Tract Surgical Unit, Veneto Institute of Oncology (IOV-IRCCS), Padova, Italy
| | - Rita Alfieri
- Esophageal and Digestive Tract Surgical Unit, Veneto Institute of Oncology (IOV-IRCCS), Padova, Italy
| | - Veronica Macchi
- Department of Molecular Medicine, Normal Anatomy Unit, University of Padova, Padova, Italy
| | - Umberto Tedeschi
- Department of General Surgery and Odontoiatrics, University Hospital of Verona, Verona, Italy
| | - Fiorella Calabrese
- Department of Cardiothoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Massimo Rugge
- Department of Medicine, Surgical Pathology & Cytopathology Unit, University of Padova, Padova, Italy
| | - Carlo Castoro
- Esophageal and Digestive Tract Surgical Unit, Veneto Institute of Oncology (IOV-IRCCS), Padova, Italy
| | - Roberto Vettor
- Department of Medicine, Endocrine-Metabolic Laboratory, University of Padova, Padova, Italy
| | - Marco Scarpa
- Esophageal and Digestive Tract Surgical Unit, Veneto Institute of Oncology (IOV-IRCCS), Padova, Italy
| |
Collapse
|
22
|
O’Halloran N, Courtney D, Kerin MJ, Lowery AJ. Adipose-Derived Stem Cells in Novel Approaches to Breast Reconstruction: Their Suitability for Tissue Engineering and Oncological Safety. Breast Cancer (Auckl) 2017; 11:1178223417726777. [PMID: 29104428 PMCID: PMC5562338 DOI: 10.1177/1178223417726777] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 07/13/2017] [Indexed: 12/13/2022] Open
Abstract
Adipose-derived stem cells (ADSCs) are rapidly becoming the gold standard cell source for tissue engineering strategies and hold great potential for novel breast reconstruction strategies. However, their use in patients with breast cancer is controversial and their oncological safety, particularly in relation to local disease recurrence, has been questioned. In vitro, in vivo, and clinical studies using ADSCs report conflicting data on their suitability for adipose tissue regeneration in patients with cancer. This review aims to provide an overview of the potential role for ADSCs in breast reconstruction and to examine the evidence relating to the oncologic safety of their use in patients with breast cancer.
Collapse
Affiliation(s)
- Niamh O’Halloran
- Discipline of Surgery, Lambe Institute for Translational Research, National University of Ireland, Galway, Galway, Ireland
| | - Donald Courtney
- Discipline of Surgery, Lambe Institute for Translational Research, National University of Ireland, Galway, Galway, Ireland
| | - Michael J Kerin
- Discipline of Surgery, Lambe Institute for Translational Research, National University of Ireland, Galway, Galway, Ireland
| | - Aoife J Lowery
- Graduate Entry Medical School, University of Limerick, Limerick, Ireland
| |
Collapse
|
23
|
Duggan C, Tapsoba JDD, Wang CY, McTiernan A. Dietary Weight Loss and Exercise Effects on Serum Biomarkers of Angiogenesis in Overweight Postmenopausal Women: A Randomized Controlled Trial. Cancer Res 2017; 76:4226-35. [PMID: 27417562 DOI: 10.1158/0008-5472.can-16-0399] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 04/28/2016] [Indexed: 01/07/2023]
Abstract
Obese and sedentary persons have an increased risk for cancer, but underlying mechanisms are poorly understood. Angiogenesis is common to adipose tissue formation and remodeling, and to tumor vascularization. A total of 439 overweight/obese, healthy, postmenopausal women [body mass index (BMI) > 25 kg/m(2)] ages 50-75 years, recruited between 2005 and 2008 were randomized to a 4-arm 12-month randomized controlled trial, comparing a caloric restriction diet arm (goal: 10% weight loss, N = 118), aerobic exercise arm (225 minutes/week of moderate-to-vigorous activity, N = 117), a combined diet + exercise arm (N = 117), or control (N = 87) on circulating levels of angiogenic biomarkers. VEGF, plasminogen activator inhibitor-1 (PAI-1), and pigment epithelium-derived factor (PEDF) were measured by immunoassay at baseline and 12 months. Changes were compared using generalized estimating equations, adjusting for baseline BMI, age, and race/ethnicity. Participants randomized to the diet + exercise arms had statistically significantly greater reductions in PAI-1 at 12 months compared with controls (-19.3% vs. +3.48%, respectively, P < 0.0001). Participants randomized to the diet and diet + exercise arms had statistically significantly greater reductions in PEDF (-9.20%, -9.90%, respectively, both P < 0.0001) and VEGF (-8.25%, P = 0.0005; -9.98%, P < 0.0001, respectively) compared with controls. There were no differences in any of the analytes in participants randomized to the exercise arm compared with controls. Increasing weight loss was statistically significantly associated with linear trends of greater reductions in PAI-1, PEDF, and VEGF. Weight loss is significantly associated with reduced circulating VEGF, PEDF, and PAI-1, and could provide incentive for reducing weight as a cancer prevention method in overweight and obese individuals. Cancer Res; 76(14); 4226-35. ©2016 AACR.
Collapse
Affiliation(s)
- Catherine Duggan
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.
| | - Jean de Dieu Tapsoba
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Ching-Yun Wang
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Anne McTiernan
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| |
Collapse
|
24
|
Pathogenic Role of IL-17-Producing Immune Cells in Obesity, and Related Inflammatory Diseases. J Clin Med 2017; 6:jcm6070068. [PMID: 28708082 PMCID: PMC5532576 DOI: 10.3390/jcm6070068] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 07/03/2017] [Accepted: 07/04/2017] [Indexed: 12/14/2022] Open
Abstract
Obesity is associated with low-grade chronic inflammation. Indeed, adipose tissues (AT) in obese individuals are the former site of progressive infiltration by pro-inflammatory immune cells, which together with increased inflammatory adipokine secretion induce adipocyte insulin resistance. IL-17-producing T (Th17) cells are part of obese AT infiltrating cells, and are likely to be promoted by adipose tissue-derived mesenchymal stem cells, as previously reported by our team. Whereas Th17 cell are physiologically implicated in the neutralization of fungal and bacterial pathogens through activation of neutrophils, they may also play a pivotal role in the onset and/or progression of chronic inflammatory diseases, or cancer, in which obesity is recognized as a risk factor. In this review, we will highlight the pathogenic role of IL-17A producing cells in the mechanisms leading to inflammation in obesity and to progression of obesity-related inflammatory diseases.
Collapse
|
25
|
Daquinag AC, Dadbin A, Snyder B, Wang X, Sahin AA, Ueno NT, Kolonin MG. Non-glycanated Decorin Is a Drug Target on Human Adipose Stromal Cells. MOLECULAR THERAPY-ONCOLYTICS 2017; 6:1-9. [PMID: 28607949 PMCID: PMC5458115 DOI: 10.1016/j.omto.2017.05.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 05/10/2017] [Indexed: 12/28/2022]
Abstract
Adipose stromal cells (ASCs) have been identified as a mesenchymal cell population recruited from white adipose tissue (WAT) by tumors and supporting cancer progression. We have previously reported the existence of a non-glycanated decorin isoform (ngDCN) marking mouse ASCs. We identified a peptide CSWKYWFGEC that binds to ngDCN and hence can serve as a vehicle for ASC-directed therapy delivery. We used hunter-killer peptides composed of CSWKYWFGEC and a pro-apoptotic moiety to deplete ASCs and suppress growth of mouse tumors. Here, we report the discovery of the human non-glycanated decorin isoform. We show that CSWKYWFGEC can be used as a probe to identify ASCs in human WAT and tumors. We demonstrate that human ngDCN is expressed on ASC surface. Finally, we validate ngDCN as a molecular target for pharmacological depletion of human ASCs with hunter-killer peptides. We propose that ngDCN-targeting agents could be developed for obesity and cancer treatment.
Collapse
Affiliation(s)
- Alexes C Daquinag
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Ali Dadbin
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Brad Snyder
- Department of Surgery, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Xiaoping Wang
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Section of Translational Breast Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Aysegul A Sahin
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Naoto T Ueno
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Section of Translational Breast Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mikhail G Kolonin
- The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
26
|
Myckatyn TM, Wagner IJ, Mehrara BJ, Crosby MA, Park JE, Qaqish BF, Moore DT, Busch EL, Silva AK, Kaur S, Ollila DW, Lee CN. Cancer Risk after Fat Transfer: A Multicenter Case-Cohort Study. Plast Reconstr Surg 2017; 139:11-18. [PMID: 28027219 PMCID: PMC5428547 DOI: 10.1097/prs.0000000000002838] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Fat transfer is an increasingly popular method for refining postmastectomy breast reconstructions. However, concern persists that fat transfer may promote disease recurrence. Adipocytes are derived from adipose-derived stem cells and express adipocytokines that can facilitate active breast cancer cells in laboratory models. The authors sought to evaluate the association between fat transfer to the reconstructed breast and cancer recurrence in patients diagnosed with local or regional invasive breast cancers. METHODS A multicenter, case-cohort study was performed. Eligible patients from four centers (Memorial Sloan Kettering, M. D. Anderson Cancer Center, Alvin J. Siteman Cancer Center, and the University of Chicago) were identified by each site's institutional tumor registry or cancer data warehouse. Eligibility criteria were as follows: mastectomy with immediate breast reconstruction between 2006 and 2011, age older than 21 years, female sex, and incident diagnosis of invasive ductal carcinoma (stage I, II, or III). Cases consisted of all recurrences during the study period, and controls consisted of a 30 percent random sample of the study population. Cox proportional hazards regression was used to evaluate for association between fat transfer and time to recurrence in bivariate and multivariate models. RESULTS The time to disease recurrence unadjusted hazard ratio for fat transfer was 0.99 (95 percent CI, 0.56 to 1.7). After adjustment for age, body mass index, stage, HER2/Neu receptor status, and estrogen receptor status, the hazard ratio was 0.97 (95 percent CI, 0.54 to 1.8). CONCLUSION In this population of breast cancer patients who had mastectomy with immediate reconstruction, fat transfer was not associated with a higher risk of cancer recurrence. CLINICAL QUESTION/LEVEL OF EVIDENCE Therapeutic, III.
Collapse
Affiliation(s)
- Terence M Myckatyn
- St. Louis, Mo.; Chapel Hill, N.C.; New York, N.Y.; Houston, Texas; Chicago and Arlington Heights, Ill.; Boston, Mass.; and Columbus, Ohio
- From the Division of Plastic and Reconstructive Surgery, Washington University School of Medicine in Saint Louis, and the Alvin J. Siteman Cancer Center; the Division of Plastic and Reconstructive Surgery, the Department of Biostatistics, the Department of Epidemiology, Gillings School of Global Public Health, the Lineberger Comprehensive Cancer Center, and the Division of Surgical Oncology, Department of Surgery, University of North Carolina; the Department of Surgery, Memorial Sloan Kettering Cancer Center; the Department of Plastic Surgery, M. D. Anderson Cancer Center; the Section of Plastic and Reconstructive Surgery, University of Chicago Medicine and Biological Sciences, University of Chicago; The Plastic Surgery Foundation; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School; the Department of Epidemiology, Harvard T. H. Chan School of Public Health; and Plastic and Reconstructive Surgery, Health Services Management and Policy, Ohio State University
| | - I Janelle Wagner
- St. Louis, Mo.; Chapel Hill, N.C.; New York, N.Y.; Houston, Texas; Chicago and Arlington Heights, Ill.; Boston, Mass.; and Columbus, Ohio
- From the Division of Plastic and Reconstructive Surgery, Washington University School of Medicine in Saint Louis, and the Alvin J. Siteman Cancer Center; the Division of Plastic and Reconstructive Surgery, the Department of Biostatistics, the Department of Epidemiology, Gillings School of Global Public Health, the Lineberger Comprehensive Cancer Center, and the Division of Surgical Oncology, Department of Surgery, University of North Carolina; the Department of Surgery, Memorial Sloan Kettering Cancer Center; the Department of Plastic Surgery, M. D. Anderson Cancer Center; the Section of Plastic and Reconstructive Surgery, University of Chicago Medicine and Biological Sciences, University of Chicago; The Plastic Surgery Foundation; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School; the Department of Epidemiology, Harvard T. H. Chan School of Public Health; and Plastic and Reconstructive Surgery, Health Services Management and Policy, Ohio State University
| | - Babak J Mehrara
- St. Louis, Mo.; Chapel Hill, N.C.; New York, N.Y.; Houston, Texas; Chicago and Arlington Heights, Ill.; Boston, Mass.; and Columbus, Ohio
- From the Division of Plastic and Reconstructive Surgery, Washington University School of Medicine in Saint Louis, and the Alvin J. Siteman Cancer Center; the Division of Plastic and Reconstructive Surgery, the Department of Biostatistics, the Department of Epidemiology, Gillings School of Global Public Health, the Lineberger Comprehensive Cancer Center, and the Division of Surgical Oncology, Department of Surgery, University of North Carolina; the Department of Surgery, Memorial Sloan Kettering Cancer Center; the Department of Plastic Surgery, M. D. Anderson Cancer Center; the Section of Plastic and Reconstructive Surgery, University of Chicago Medicine and Biological Sciences, University of Chicago; The Plastic Surgery Foundation; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School; the Department of Epidemiology, Harvard T. H. Chan School of Public Health; and Plastic and Reconstructive Surgery, Health Services Management and Policy, Ohio State University
| | - Melissa A Crosby
- St. Louis, Mo.; Chapel Hill, N.C.; New York, N.Y.; Houston, Texas; Chicago and Arlington Heights, Ill.; Boston, Mass.; and Columbus, Ohio
- From the Division of Plastic and Reconstructive Surgery, Washington University School of Medicine in Saint Louis, and the Alvin J. Siteman Cancer Center; the Division of Plastic and Reconstructive Surgery, the Department of Biostatistics, the Department of Epidemiology, Gillings School of Global Public Health, the Lineberger Comprehensive Cancer Center, and the Division of Surgical Oncology, Department of Surgery, University of North Carolina; the Department of Surgery, Memorial Sloan Kettering Cancer Center; the Department of Plastic Surgery, M. D. Anderson Cancer Center; the Section of Plastic and Reconstructive Surgery, University of Chicago Medicine and Biological Sciences, University of Chicago; The Plastic Surgery Foundation; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School; the Department of Epidemiology, Harvard T. H. Chan School of Public Health; and Plastic and Reconstructive Surgery, Health Services Management and Policy, Ohio State University
| | - Julie E Park
- St. Louis, Mo.; Chapel Hill, N.C.; New York, N.Y.; Houston, Texas; Chicago and Arlington Heights, Ill.; Boston, Mass.; and Columbus, Ohio
- From the Division of Plastic and Reconstructive Surgery, Washington University School of Medicine in Saint Louis, and the Alvin J. Siteman Cancer Center; the Division of Plastic and Reconstructive Surgery, the Department of Biostatistics, the Department of Epidemiology, Gillings School of Global Public Health, the Lineberger Comprehensive Cancer Center, and the Division of Surgical Oncology, Department of Surgery, University of North Carolina; the Department of Surgery, Memorial Sloan Kettering Cancer Center; the Department of Plastic Surgery, M. D. Anderson Cancer Center; the Section of Plastic and Reconstructive Surgery, University of Chicago Medicine and Biological Sciences, University of Chicago; The Plastic Surgery Foundation; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School; the Department of Epidemiology, Harvard T. H. Chan School of Public Health; and Plastic and Reconstructive Surgery, Health Services Management and Policy, Ohio State University
| | - Bahjat F Qaqish
- St. Louis, Mo.; Chapel Hill, N.C.; New York, N.Y.; Houston, Texas; Chicago and Arlington Heights, Ill.; Boston, Mass.; and Columbus, Ohio
- From the Division of Plastic and Reconstructive Surgery, Washington University School of Medicine in Saint Louis, and the Alvin J. Siteman Cancer Center; the Division of Plastic and Reconstructive Surgery, the Department of Biostatistics, the Department of Epidemiology, Gillings School of Global Public Health, the Lineberger Comprehensive Cancer Center, and the Division of Surgical Oncology, Department of Surgery, University of North Carolina; the Department of Surgery, Memorial Sloan Kettering Cancer Center; the Department of Plastic Surgery, M. D. Anderson Cancer Center; the Section of Plastic and Reconstructive Surgery, University of Chicago Medicine and Biological Sciences, University of Chicago; The Plastic Surgery Foundation; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School; the Department of Epidemiology, Harvard T. H. Chan School of Public Health; and Plastic and Reconstructive Surgery, Health Services Management and Policy, Ohio State University
| | - Dominic T Moore
- St. Louis, Mo.; Chapel Hill, N.C.; New York, N.Y.; Houston, Texas; Chicago and Arlington Heights, Ill.; Boston, Mass.; and Columbus, Ohio
- From the Division of Plastic and Reconstructive Surgery, Washington University School of Medicine in Saint Louis, and the Alvin J. Siteman Cancer Center; the Division of Plastic and Reconstructive Surgery, the Department of Biostatistics, the Department of Epidemiology, Gillings School of Global Public Health, the Lineberger Comprehensive Cancer Center, and the Division of Surgical Oncology, Department of Surgery, University of North Carolina; the Department of Surgery, Memorial Sloan Kettering Cancer Center; the Department of Plastic Surgery, M. D. Anderson Cancer Center; the Section of Plastic and Reconstructive Surgery, University of Chicago Medicine and Biological Sciences, University of Chicago; The Plastic Surgery Foundation; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School; the Department of Epidemiology, Harvard T. H. Chan School of Public Health; and Plastic and Reconstructive Surgery, Health Services Management and Policy, Ohio State University
| | - Evan L Busch
- St. Louis, Mo.; Chapel Hill, N.C.; New York, N.Y.; Houston, Texas; Chicago and Arlington Heights, Ill.; Boston, Mass.; and Columbus, Ohio
- From the Division of Plastic and Reconstructive Surgery, Washington University School of Medicine in Saint Louis, and the Alvin J. Siteman Cancer Center; the Division of Plastic and Reconstructive Surgery, the Department of Biostatistics, the Department of Epidemiology, Gillings School of Global Public Health, the Lineberger Comprehensive Cancer Center, and the Division of Surgical Oncology, Department of Surgery, University of North Carolina; the Department of Surgery, Memorial Sloan Kettering Cancer Center; the Department of Plastic Surgery, M. D. Anderson Cancer Center; the Section of Plastic and Reconstructive Surgery, University of Chicago Medicine and Biological Sciences, University of Chicago; The Plastic Surgery Foundation; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School; the Department of Epidemiology, Harvard T. H. Chan School of Public Health; and Plastic and Reconstructive Surgery, Health Services Management and Policy, Ohio State University
| | - Amanda K Silva
- St. Louis, Mo.; Chapel Hill, N.C.; New York, N.Y.; Houston, Texas; Chicago and Arlington Heights, Ill.; Boston, Mass.; and Columbus, Ohio
- From the Division of Plastic and Reconstructive Surgery, Washington University School of Medicine in Saint Louis, and the Alvin J. Siteman Cancer Center; the Division of Plastic and Reconstructive Surgery, the Department of Biostatistics, the Department of Epidemiology, Gillings School of Global Public Health, the Lineberger Comprehensive Cancer Center, and the Division of Surgical Oncology, Department of Surgery, University of North Carolina; the Department of Surgery, Memorial Sloan Kettering Cancer Center; the Department of Plastic Surgery, M. D. Anderson Cancer Center; the Section of Plastic and Reconstructive Surgery, University of Chicago Medicine and Biological Sciences, University of Chicago; The Plastic Surgery Foundation; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School; the Department of Epidemiology, Harvard T. H. Chan School of Public Health; and Plastic and Reconstructive Surgery, Health Services Management and Policy, Ohio State University
| | - Surinder Kaur
- St. Louis, Mo.; Chapel Hill, N.C.; New York, N.Y.; Houston, Texas; Chicago and Arlington Heights, Ill.; Boston, Mass.; and Columbus, Ohio
- From the Division of Plastic and Reconstructive Surgery, Washington University School of Medicine in Saint Louis, and the Alvin J. Siteman Cancer Center; the Division of Plastic and Reconstructive Surgery, the Department of Biostatistics, the Department of Epidemiology, Gillings School of Global Public Health, the Lineberger Comprehensive Cancer Center, and the Division of Surgical Oncology, Department of Surgery, University of North Carolina; the Department of Surgery, Memorial Sloan Kettering Cancer Center; the Department of Plastic Surgery, M. D. Anderson Cancer Center; the Section of Plastic and Reconstructive Surgery, University of Chicago Medicine and Biological Sciences, University of Chicago; The Plastic Surgery Foundation; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School; the Department of Epidemiology, Harvard T. H. Chan School of Public Health; and Plastic and Reconstructive Surgery, Health Services Management and Policy, Ohio State University
| | - David W Ollila
- St. Louis, Mo.; Chapel Hill, N.C.; New York, N.Y.; Houston, Texas; Chicago and Arlington Heights, Ill.; Boston, Mass.; and Columbus, Ohio
- From the Division of Plastic and Reconstructive Surgery, Washington University School of Medicine in Saint Louis, and the Alvin J. Siteman Cancer Center; the Division of Plastic and Reconstructive Surgery, the Department of Biostatistics, the Department of Epidemiology, Gillings School of Global Public Health, the Lineberger Comprehensive Cancer Center, and the Division of Surgical Oncology, Department of Surgery, University of North Carolina; the Department of Surgery, Memorial Sloan Kettering Cancer Center; the Department of Plastic Surgery, M. D. Anderson Cancer Center; the Section of Plastic and Reconstructive Surgery, University of Chicago Medicine and Biological Sciences, University of Chicago; The Plastic Surgery Foundation; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School; the Department of Epidemiology, Harvard T. H. Chan School of Public Health; and Plastic and Reconstructive Surgery, Health Services Management and Policy, Ohio State University
| | - Clara N Lee
- St. Louis, Mo.; Chapel Hill, N.C.; New York, N.Y.; Houston, Texas; Chicago and Arlington Heights, Ill.; Boston, Mass.; and Columbus, Ohio
- From the Division of Plastic and Reconstructive Surgery, Washington University School of Medicine in Saint Louis, and the Alvin J. Siteman Cancer Center; the Division of Plastic and Reconstructive Surgery, the Department of Biostatistics, the Department of Epidemiology, Gillings School of Global Public Health, the Lineberger Comprehensive Cancer Center, and the Division of Surgical Oncology, Department of Surgery, University of North Carolina; the Department of Surgery, Memorial Sloan Kettering Cancer Center; the Department of Plastic Surgery, M. D. Anderson Cancer Center; the Section of Plastic and Reconstructive Surgery, University of Chicago Medicine and Biological Sciences, University of Chicago; The Plastic Surgery Foundation; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School; the Department of Epidemiology, Harvard T. H. Chan School of Public Health; and Plastic and Reconstructive Surgery, Health Services Management and Policy, Ohio State University
| |
Collapse
|
27
|
Divella R, De Luca R, Abbate I, Naglieri E, Daniele A. Obesity and cancer: the role of adipose tissue and adipo-cytokines-induced chronic inflammation. J Cancer 2016; 7:2346-2359. [PMID: 27994674 PMCID: PMC5166547 DOI: 10.7150/jca.16884] [Citation(s) in RCA: 204] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 09/19/2016] [Indexed: 12/12/2022] Open
Abstract
Adipose tissue in addition to its ability to keep lipids is now recognized as a real organ with both metabolic and endocrine functions. Recent studies demonstrated that in obese animals is established a status of adipocyte hypoxia and in this hypoxic state interaction between adipocytes and stromal vascular cells contribute to tumor development and progression. In several tumors such as breast, colon, liver and prostate, obesity represents a poor predictor of clinical outcomes. Dysfunctional adipose tissue in obesity releases a disturbed profile of adipokines with elevated levels of pro-inflammatory factors and a consequent alteration of key signaling mediators which may be an active local player in establishing the peritumoral environment promoting tumor growth and progression. Therefore, adipose tissue hypoxia might contribute to cancer risk in the obese population. To date the precise mechanisms behind this obesity-cancer link is not yet fully understood. In the light of information provided in this review that aims to identify the key mechanisms underlying the link between obesity and cancer we support that inflammatory state specific of obesity may be important in obesity-cancer link.
Collapse
Affiliation(s)
- Rosa Divella
- Clinical Pathology Laboratory, Department of Experimental Oncology. Giovanni Paolo II National Cancer Institute, V.Le Orazio Flacco 65, 70124 -Bari, Italy
| | - Raffaele De Luca
- Department of Surgery Oncology. Giovanni Paolo II National Cancer Institute, V.Le Orazio Flacco 65, 70124 -Bari, Italy
| | - Ines Abbate
- Clinical Pathology Laboratory, Department of Experimental Oncology. Giovanni Paolo II National Cancer Institute, V.Le Orazio Flacco 65, 70124 -Bari, Italy
| | - Emanuele Naglieri
- Department of Medical Oncology, Giovanni Paolo II National Cancer Institute, V.Le Orazio Flacco 65, 70124 -Bari, Italy
| | - Antonella Daniele
- Clinical Pathology Laboratory, Department of Experimental Oncology. Giovanni Paolo II National Cancer Institute, V.Le Orazio Flacco 65, 70124 -Bari, Italy
| |
Collapse
|
28
|
Waked K, Colle J, Doornaert M, Cocquyt V, Blondeel P. Systematic review: The oncological safety of adipose fat transfer after breast cancer surgery. Breast 2016; 31:128-136. [PMID: 27837706 DOI: 10.1016/j.breast.2016.11.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 10/02/2016] [Accepted: 11/03/2016] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVES Oncological concerns have risen around the safety of adipose fat transfer (AFT) after breast cancer surgery. In this article, we present the clinical and molecular evidences, and discuss the current contradiction between them. MATERIALS AND METHODS Every clinical trial and experimental study on AFT and its oncological influences was screened. Between September 2014 and September 2016, 856 articles from four databases were found. 105 core articles were selected. RESULTS A total of 18 clinical studies have been published. The loco-regional recurrence (LRR) incidence rates range between 0 and 3.90% per year. For the mastectomy and breast conservative therapy group separately, a LRR per year between 0 and 1.62% and 0-3.90 has been reported, respectively. Some studies included a matched control group and found no significant difference between cases and controls, with the exception of a subgroup of patients with intraepithelial breast carcinoma. Adipose derived mesenchymal stem cells have a potential oncogenic effect on residual cancer cells after breast cancer surgery. Numerous signalling proteins and pathways have been described that can stimulate tumour initiation and growth. CONCLUSION There is a contradiction between experimental and clinical findings. Numerous adipokines have been discovered that could potentially promote tumour initiation and growth, but clinical studies fail to point out a significant increase in LRR in patients who receive AFT after breast cancer surgery. More prospective studies are needed with a sufficient follow-up time and analysis of some critical factors, such as adjuvant radiotherapy and hormonal therapy, the origin and volume of the injected fat, and genetic influences.
Collapse
Affiliation(s)
- Karl Waked
- University Hospital of Ghent, De Pintelaan 185, 9000 Gent, Belgium.
| | - Julien Colle
- University Hospital of Ghent, De Pintelaan 185, 9000 Gent, Belgium.
| | - Maarten Doornaert
- Private Medical Center: Maaltebrugge Ghent, Maaltebruggestraat 288, 9000 Gent, Belgium.
| | | | - Phillip Blondeel
- University Hospital of Ghent, De Pintelaan 185, 9000 Gent, Belgium.
| |
Collapse
|
29
|
Zhang Y, Kolonin MG. Cytokine signaling regulating adipose stromal cell trafficking. Adipocyte 2016; 5:369-374. [PMID: 27994950 DOI: 10.1080/21623945.2016.1220452] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 07/27/2016] [Accepted: 07/28/2016] [Indexed: 01/31/2023] Open
Abstract
Adipocyte progenitors, known as adipose stromal cells (ASC), can become mobilized, recruited by tumors, and contribute to cancer progression. Mechanisms underlying ASC trafficking have remained obscure. We recently reported that CXCL1 expressed by cancer cells chemoattracts ASC expressing CXCR1 in obesity. As a candidate mechanism of CXCL1 activation, we identified interleukin (IL)-22, systemic circulation of which is increased in obesity. It has been reported that IL-22 signaling through IL-22R is upstream of CXCL1. Here, we provide evidence that IL-22 expression by leukocytes infiltrating WAT and IL-22R expression by tumors is obesity-dependent. We propose that obesity-associated adipocyte death and the resulting recruitment of leukocytes triggers the IL-22 signaling cascade that induces CXCL1 secretion by cancer cells responsible for ASC trafficking to tumors.
Collapse
|
30
|
Zhang Y, Ertl HCJ. Starved and Asphyxiated: How Can CD8(+) T Cells within a Tumor Microenvironment Prevent Tumor Progression. Front Immunol 2016; 7:32. [PMID: 26904023 PMCID: PMC4748049 DOI: 10.3389/fimmu.2016.00032] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 01/22/2016] [Indexed: 01/08/2023] Open
Abstract
Although cancer immunotherapy has achieved significant breakthroughs in recent years, its overall efficacy remains limited in the majority of patients. One major barrier is exhaustion of tumor antigen-specific CD8(+) tumor-infiltrating lymphocytes (TILs), which conventionally has been attributed to persistent stimulation with antigen within the tumor microenvironment (TME). A series of recent studies have highlighted that the TME poses significant metabolic challenges to TILs, which may contribute to their functional exhaustion. Hypoxia increases the expression of coinhibitors on activated CD8(+) T cells, which in general reduces the T cells' effector functions. It also impairs the cells' ability to gain energy through oxidative phosphorylation. Glucose limitation increases the expression of programed cell death protein-1 and reduces functions of activated CD8(+) T cells. A combination of hypoxia and hypoglycemia, as is common in solid tumors, places CD8(+) TILs at dual metabolic jeopardy by affecting both major pathways of energy production. Recently, a number of studies addressed the effects of metabolic stress on modulating CD8(+) T cell metabolism, differentiation, and functions. Here, we discuss recent findings on how different types of metabolic stress within the TME shape the tumor-killing capacity of CD8(+) T cells. We propose that manipulating the metabolism of TILs to more efficiently utilize nutrients, especially during intermittent periods of hypoxia could maximize their performance, prolong their survival and improve the efficacy of active cancer immunotherapy.
Collapse
Affiliation(s)
- Ying Zhang
- Gene Therapy and Vaccines Program, University of Pennsylvania School of Medicine, Philadelphia, PA, USA; The Wistar Institute Vaccine Center, Philadelphia, PA, USA
| | | |
Collapse
|
31
|
Daley-Brown D, Oprea-Ilies GM, Lee R, Pattillo R, Gonzalez-Perez RR. Molecular cues on obesity signals, tumor markers and endometrial cancer. Horm Mol Biol Clin Investig 2015; 21:89-106. [PMID: 25781554 DOI: 10.1515/hmbci-2014-0049] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 01/28/2015] [Indexed: 02/07/2023]
Abstract
Tumor markers are important tools for early diagnosis, prognosis, therapy response and endometrial cancer monitoring. A large number of molecular and pathologic markers have been described in types I and II endometrial cancers, which has served to define the main oncogenic, epidemiological, genetic, clinical and histopathological features. Ongoing attempts to stratify biological markers of endometrial cancer are presented. However, data on changes in tumor marker profiles in obesity-related endometrial cancer are scarce. Obesity is a pandemic in Western countries that has an important impact on endometrial cancers, albeit through not very well-defined mechanisms. Although endometrial cancer is more common in Caucasian women, higher mortality is found in African Americans who also show higher incidence of obesity. Here, we describe how obesity signals (estrogen, leptin, leptin induced-molecules, Notch; cytokines and growth factors) could affect endometrial cancer. Leptin signaling and its crosstalk may be associated to the more aggressive and poor prognosis type II endometrial cancer, which affects more postmenopausal and African-American women. In this regard, studies on expression of novel molecular markers (Notch, interleukin-1 and leptin crosstalk outcome) may provide essential clues for detection, prevention, treatment and prognosis.
Collapse
|
32
|
Abstract
PURPOSE OF REVIEW There is emerging evidence that obesity is associated with an increase in the incidence, severity, and mortality from different types of cancer, including postmenopausal breast cancer. Here, we discuss the role of white adipose tissue (WAT) cells and of related soluble factors in the local and metastatic growth of this neoplastic disease. Moreover, we discuss the recent increase in the use of WAT-derived progenitor cells in breast cancer patients to enhance the quality of breast reconstruction and the related risks. RECENT FINDINGS In several murine models, WAT cells and progenitors were found to have cooperative roles in promoting local breast cancer. Moreover, they were found to contribute to adipocytes and pericytes supporting the cancer vasculature, and stimulated the metastatic progression of breast cancer. There are |