1
|
Vickers RR, Wyatt GL, Sanchez L, VanPortfliet JJ, West AP, Porter WW. Loss of STING impairs lactogenic differentiation. Development 2024; 151:dev202998. [PMID: 39399905 PMCID: PMC11528151 DOI: 10.1242/dev.202998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 08/27/2024] [Indexed: 10/15/2024]
Abstract
Heightened energetic and nutrient demand during lactogenic differentiation of the mammary gland elicits upregulation of various stress responses to support cellular homeostasis. Here, we identify the stimulator of interferon genes (STING) as an immune supporter of the functional development of mouse mammary epithelial cells (MECs). An in vitro model of MEC differentiation revealed that STING is activated in a cGAS-independent manner to produce both type I interferons and proinflammatory cytokines in response to the accumulation of mitochondrial reactive oxygen species. Induction of STING activity was found to be dependent on the breast tumor suppressor gene single-minded 2 (SIM2). Using mouse models of lactation, we discovered that loss of STING activity results in early involution of #3 mammary glands, severely impairing lactational performance. Our data suggest that STING is required for successful functional differentiation of the mammary gland and bestows a differential lactogenic phenotype between #3 mammary glands and the traditionally explored inguinal 4|9 pair. These findings affirm unique development of mammary gland pairs that is essential to consider in future investigations into normal development and breast cancer initiation.
Collapse
Affiliation(s)
- Ramiah R. Vickers
- Department of Veterinary Physiology & Pharmacology, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843, USA
| | - Garhett L. Wyatt
- Department of Veterinary Physiology & Pharmacology, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843, USA
| | - Lilia Sanchez
- Department of Veterinary Physiology & Pharmacology, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843, USA
| | | | | | - Weston W. Porter
- Department of Veterinary Physiology & Pharmacology, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
2
|
Ye DM, Bai X, Xu S, Qu N, Zhao N, Zheng Y, Yu T, Wu H. Association between breastfeeding, mammographic density, and breast cancer risk: a review. Int Breastfeed J 2024; 19:65. [PMID: 39285438 PMCID: PMC11406879 DOI: 10.1186/s13006-024-00672-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 09/07/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Mammographic density has been associated with breast cancer risk, and is modulated by established breast cancer risk factors, such as reproductive and hormonal history, as well as lifestyle. Recent epidemiological and biological findings underscore the recognized benefits of breastfeeding in reducing breast cancer risk, especially for aggressive subtypes. Current research exploring the association among mammographic density, breastfeeding, and breast cancer is sparse. MAIN FINDINGS Changes occur in the breasts during pregnancy in preparation for lactation, characterized by the proliferation of mammary gland tissues and the development of mammary alveoli. During lactation, the alveoli fill with milk, and subsequent weaning triggers the involution and remodeling of these tissues. Breastfeeding influences the breast microenvironment, potentially altering mammographic density. When breastfeeding is not initiated after birth, or is abruptly discontinued shortly after, the breast tissue undergoes forced and abrupt involution. Conversely, when breastfeeding is sustained over an extended period and concludes gradually, the breast tissue undergoes slow remodeling process known as gradual involution. Breast tissue undergoing abrupt involution displays denser stroma, altered collagen composition, heightened inflammation and proliferation, along with increased expression of estrogen receptor α (ERα) and progesterone receptor. Furthermore, elevated levels of pregnancy-associated plasma protein-A (PAPP-A) surpass those of its inhibitors during abrupt involution, enhancing insulin-like growth factor (IGF) signaling and collagen deposition. Prolactin and small molecules in breast milk may also modulate DNA methylation levels. Drawing insights from contemporary epidemiological and molecular biology studies, our review sheds light on how breastfeeding impacts mammographic density and explores its role in influencing breast cancer. CONCLUSION This review highlights a clear protective link between breastfeeding and reduced breast cancer risk via changes in mammographic density. Future research should investigate the effects of breastfeeding on mammographic density and breast cancer risk among various ethnic groups and elucidate the molecular mechanisms underlying these associations. Such comprehensive research will enhance our understanding and facilitate the development of targeted breast cancer prevention and treatment strategies.
Collapse
Affiliation(s)
- Dong-Man Ye
- Department of Medical Imaging, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China
| | - Xiaoru Bai
- Department of Medical Imaging, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China
| | - Shu Xu
- Department of Medical Imaging, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China
| | - Ning Qu
- Department of Medical Imaging, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China
| | - Nannan Zhao
- Department of Medical Imaging, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China
| | - Yang Zheng
- Department of Laboratory Medicine, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China
| | - Tao Yu
- Department of Medical Imaging, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China.
| | - Huijian Wu
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, 116024, Liaoning Province, China.
| |
Collapse
|
3
|
Zhang H, Felthaus O, Eigenberger A, Klein S, Prantl L. Treg Cell Therapeutic Strategies for Breast Cancer: Holistic to Local Aspects. Cells 2024; 13:1526. [PMID: 39329710 PMCID: PMC11429654 DOI: 10.3390/cells13181526] [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: 07/30/2024] [Revised: 09/06/2024] [Accepted: 09/10/2024] [Indexed: 09/28/2024] Open
Abstract
Regulatory T cells (Tregs) play a key role in maintaining immune homeostasis and preventing autoimmunity through their immunosuppressive function. There have been numerous reports confirming that high levels of Tregs in the tumor microenvironment (TME) are associated with a poor prognosis, highlighting their role in promoting an immunosuppressive environment. In breast cancer (BC), Tregs interact with cancer cells, ultimately leading to the suppression of immune surveillance and promoting tumor progression. This review discusses the dual role of Tregs in breast cancer, and explores the controversies and therapeutic potential associated with targeting these cells. Researchers are investigating various strategies to deplete or inhibit Tregs, such as immune checkpoint inhibitors, cytokine antagonists, and metabolic inhibition. However, the heterogeneity of Tregs and the variable precision of treatments pose significant challenges. Understanding the functional diversity of Tregs and the latest advances in targeted therapies is critical for the development of effective therapies. This review highlights the latest approaches to Tregs for BC treatment that both attenuate Treg-mediated immunosuppression in tumors and maintain immune tolerance, and advocates precise combination therapy strategies to optimize breast cancer outcomes.
Collapse
Affiliation(s)
- Hanwen Zhang
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz-Josef-Strauss Allee 11, 93053 Regensburg, Germany (L.P.)
| | | | | | | | | |
Collapse
|
4
|
Vickers R, Porter W. Immune Cell Contribution to Mammary Gland Development. J Mammary Gland Biol Neoplasia 2024; 29:16. [PMID: 39177859 PMCID: PMC11343902 DOI: 10.1007/s10911-024-09568-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 06/27/2024] [Indexed: 08/24/2024] Open
Abstract
Postpartum breast cancer (PPBC) is a unique subset of breast cancer, accounting for nearly half of the women diagnosed during their postpartum years. Mammary gland involution is widely regarded as being a key orchestrator in the initiation and progression of PPBC due to its unique wound-healing inflammatory signature. Here, we provide dialogue suggestive that lactation may also facilitate neoplastic development as a result of sterile inflammation. Immune cells are involved in all stages of postnatal mammary development. It has been proposed that the functions of these immune cells are partially directed by mammary epithelial cells (MECs) and the cytokines they produce. This suggests that a more niche area of exploration aimed at assessing activation of innate immune pathways within MECs could provide insight into immune cell contributions to the developing mammary gland. Immune cell contribution to pubertal development and mammary gland involution has been extensively studied; however, investigations into pregnancy and lactation remain limited. During pregnancy, the mammary gland undergoes dramatic expansion to prepare for lactation. As a result, MECs are susceptible to replicative stress. During lactation, mitochondria are pushed to capacity to fulfill the high energetic demands of producing milk. This replicative and metabolic stress, if unresolved, can elicit activation of innate immune pathways within differentiating MECs. In this review, we broadly discuss postnatal mammary development and current knowledge of immune cell contribution to each developmental stage, while also emphasizing a more unique area of study that will be beneficial in the discovery of novel therapeutic biomarkers of PPBC.
Collapse
Affiliation(s)
- Ramiah Vickers
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine, Texas A&M University, College Station, TX, 77843, USA
| | - Weston Porter
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine, Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
5
|
Zhao J, Zhang K, Sui D, Wang S, Li Y, Tang X, Liu X, Song Y, Deng Y. Recent advances in sialic acid-based active targeting chemoimmunotherapy promoting tumor shedding: a systematic review. NANOSCALE 2024; 16:14621-14639. [PMID: 39023195 DOI: 10.1039/d4nr01740d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Tumors have always been a major public health concern worldwide, and attempts to look for effective treatments have never ceased. Sialic acid is known to be a crucial element for tumor development and its receptors are highly expressed on tumor-associated immune cells, which perform significant roles in establishing the immunosuppressive tumor microenvironment and further boosting tumorigenesis, progression, and metastasis. Obviously, it is essential to consider sophisticated crosstalk between tumors, the immune system, and preparations, and understand the links between pharmaceutics and immunology. Sialic acid-based chemoimmunotherapy enables active targeting drug delivery via mediating the recognition between the sialic acid-modified nano-drug delivery system represented by liposomes and sialic acid-binding receptors on tumor-associated immune cells, which inhibit their activity and utilize their homing ability to deliver drugs. Such a "Trojan horse" strategy has remarkably improved the shortcomings of traditional passive targeting treatments, unexpectedly promoted tumor shedding, and persistently induced robust immunological memory, thus highlighting its prospective application potential for targeting various tumors. Herein, we review recent advances in sialic acid-based active targeting chemoimmunotherapy to promote tumor shedding, summarize the current viewpoints on the tumor shedding mechanism, especially the formation of durable immunological memory, and analyze the challenges and opportunities of this attractive approach.
Collapse
Affiliation(s)
- Jingyi Zhao
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China.
| | - Kunfeng Zhang
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China.
| | - Dezhi Sui
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China.
| | - Shuo Wang
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China.
| | - Yantong Li
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China.
| | - Xueying Tang
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China.
| | - Xinrong Liu
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China.
| | - Yanzhi Song
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China.
| | - Yihui Deng
- College of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road, No. 103, Shenyang 110016, China.
| |
Collapse
|
6
|
Zhou Y, Meyle J, Groeger S. Periodontal pathogens and cancer development. Periodontol 2000 2024. [PMID: 38965193 DOI: 10.1111/prd.12590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 05/03/2024] [Accepted: 06/11/2024] [Indexed: 07/06/2024]
Abstract
Increasing evidence suggests a significant association between periodontal disease and the occurrence of various cancers. The carcinogenic potential of several periodontal pathogens has been substantiated in vitro and in vivo. This review provides a comprehensive overview of the diverse mechanisms employed by different periodontal pathogens in the development of cancer. These mechanisms induce chronic inflammation, inhibit the host's immune system, activate cell invasion and proliferation, possess anti-apoptotic activity, and produce carcinogenic substances. Elucidating these mechanisms might provide new insights for developing novel approaches for tumor prevention, therapeutic purposes, and survival improvement.
Collapse
Affiliation(s)
- Yuxi Zhou
- Department of Periodontology, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Joerg Meyle
- Department of Periodontology, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Sabine Groeger
- Department of Periodontology, Justus-Liebig-University of Giessen, Giessen, Germany
- Department of Orthodontics, Justus-Liebig-University of Giessen, Giessen, Germany
| |
Collapse
|
7
|
Ruocco MR, Gisonna A, Acampora V, D’Agostino A, Carrese B, Santoro J, Venuta A, Nasso R, Rocco N, Russo D, Cavaliere A, Altobelli GG, Masone S, Avagliano A, Arcucci A, Fiume G. Guardians and Mediators of Metastasis: Exploring T Lymphocytes, Myeloid-Derived Suppressor Cells, and Tumor-Associated Macrophages in the Breast Cancer Microenvironment. Int J Mol Sci 2024; 25:6224. [PMID: 38892411 PMCID: PMC11172575 DOI: 10.3390/ijms25116224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/30/2024] [Accepted: 06/01/2024] [Indexed: 06/21/2024] Open
Abstract
Breast cancers (BCs) are solid tumors composed of heterogeneous tissues consisting of cancer cells and an ever-changing tumor microenvironment (TME). The TME includes, among other non-cancer cell types, immune cells influencing the immune context of cancer tissues. In particular, the cross talk of immune cells and their interactions with cancer cells dramatically influence BC dissemination, immunoediting, and the outcomes of cancer therapies. Tumor-infiltrating lymphocytes (TILs), tumor-associated macrophages (TAMs), and myeloid-derived suppressor cells (MDSCs) represent prominent immune cell populations of breast TMEs, and they have important roles in cancer immunoescape and dissemination. Therefore, in this article we review the features of TILs, TAMs, and MDSCs in BCs. Moreover, we highlight the mechanisms by which these immune cells remodel the immune TME and lead to breast cancer metastasis.
Collapse
Affiliation(s)
- Maria Rosaria Ruocco
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (M.R.R.); (A.G.)
| | - Armando Gisonna
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; (M.R.R.); (A.G.)
| | - Vittoria Acampora
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (V.A.); (A.V.); (A.A.)
| | - Anna D’Agostino
- IRCCS SYNLAB SDN, Via Emanuele Gianturco 113, 80143 Naples, Italy; (A.D.); (B.C.); (J.S.)
| | - Barbara Carrese
- IRCCS SYNLAB SDN, Via Emanuele Gianturco 113, 80143 Naples, Italy; (A.D.); (B.C.); (J.S.)
| | - Jessie Santoro
- IRCCS SYNLAB SDN, Via Emanuele Gianturco 113, 80143 Naples, Italy; (A.D.); (B.C.); (J.S.)
| | - Alessandro Venuta
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (V.A.); (A.V.); (A.A.)
| | - Rosarita Nasso
- Department of Movement Sciences and Wellness, University of Naples “Parthenope”, 80133 Naples, Italy;
| | - Nicola Rocco
- Department of Advanced Biomedical Science, University of Naples Federico II, 80131 Naples, Italy; (N.R.); (D.R.); (G.G.A.)
| | - Daniela Russo
- Department of Advanced Biomedical Science, University of Naples Federico II, 80131 Naples, Italy; (N.R.); (D.R.); (G.G.A.)
| | | | - Giovanna Giuseppina Altobelli
- Department of Advanced Biomedical Science, University of Naples Federico II, 80131 Naples, Italy; (N.R.); (D.R.); (G.G.A.)
| | - Stefania Masone
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy;
| | - Angelica Avagliano
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (V.A.); (A.V.); (A.A.)
| | - Alessandro Arcucci
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (V.A.); (A.V.); (A.A.)
| | - Giuseppe Fiume
- Department of Experimental and Clinical Medicine, University of Catanzaro “Magna Graecia”, 88100 Catanzaro, Italy;
| |
Collapse
|
8
|
Ivanova E, Hue-Beauvais C, Castille J, Laubier J, Le Guillou S, Aujean E, Lecardonnel J, Lebrun L, Jaffrezic F, Rousseau-Ralliard D, Péchoux C, Letheule M, Foucras G, Charlier M, Le Provost F. Mutation of SOCS2 induces structural and functional changes in mammary development. Development 2024; 151:dev202332. [PMID: 38391249 DOI: 10.1242/dev.202332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 02/15/2024] [Indexed: 02/24/2024]
Abstract
Lactation is an essential process for mammals. In sheep, the R96C mutation in suppressor of cytokine signaling 2 (SOCS2) protein is associated with greater milk production and increased mastitis sensitivity. To shed light on the involvement of R96C mutation in mammary gland development and lactation, we developed a mouse model carrying this mutation (SOCS2KI/KI). Mammary glands from virgin adult SOCS2KI/KI mice presented a branching defect and less epithelial tissue, which were not compensated for in later stages of mammary development. Mammary epithelial cell (MEC) subpopulations were modified, with mutated mice having three times as many basal cells, accompanied by a decrease in luminal cells. The SOCS2KI/KI mammary gland remained functional; however, MECs contained more lipid droplets versus fat globules, and milk lipid composition was modified. Moreover, the gene expression dynamic from virgin to pregnancy state resulted in the identification of about 3000 differentially expressed genes specific to SOCS2KI/KI or control mice. Our results show that SOCS2 is important for mammary gland development and milk production. In the long term, this finding raises the possibility of ensuring adequate milk production without compromising animal health and welfare.
Collapse
Affiliation(s)
- Elitsa Ivanova
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas 78350, France
| | - Cathy Hue-Beauvais
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas 78350, France
| | - Johan Castille
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas 78350, France
| | - Johann Laubier
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas 78350, France
| | - Sandrine Le Guillou
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas 78350, France
| | - Etienne Aujean
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas 78350, France
| | - Jerome Lecardonnel
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas 78350, France
| | - Laura Lebrun
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas 78350, France
| | - Florence Jaffrezic
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas 78350, France
| | - Delphine Rousseau-Ralliard
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas 78350, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort 94700, France
| | - Christine Péchoux
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas 78350, France
| | - Martine Letheule
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas 78350, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort 94700, France
| | - Gilles Foucras
- IHAP, Université de Toulouse, INRAE, ENVT, Toulouse 31076, France
| | - Madia Charlier
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas 78350, France
| | - Fabienne Le Provost
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas 78350, France
| |
Collapse
|
9
|
Sinha S, Hembram KC, Chatterjee S. Targeting signaling pathways in cancer stem cells: A potential approach for developing novel anti-cancer therapeutics. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 385:157-209. [PMID: 38663959 DOI: 10.1016/bs.ircmb.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Cancer stem cells (CSCs) have emerged as prime players in the intricate landscape of cancer development, progression, and resistance to traditional treatments. These unique cellular subpopulations own the remarkable capability of self-renewal and differentiation, giving rise to the diverse cellular makeup of tumors and fostering their recurrence following conventional therapies. In the quest for developing more effective cancer therapeutics, the focus has now shifted toward targeting the signaling pathways that govern CSCs behavior. This chapter underscores the significance of these signaling pathways in CSC biology and their potential as pivotal targets for the development of novel chemotherapy approaches. We delve into several key signaling pathways essential for maintaining the defining characteristics of CSCs, including the Wnt, Hedgehog, Notch, JAK-STAT, NF-κB pathways, among others, shedding light on their potential crosstalk. Furthermore, we highlight the latest advancements in CSC-targeted therapies, spanning from promising preclinical models to ongoing clinical trials. A comprehensive understanding of the intricate molecular aspects of CSC signaling pathways and their manipulation holds the prospective to revolutionize cancer treatment paradigms. This, in turn, could lead to more efficacious and personalized therapies with the ultimate goal of eradicating CSCs and enhancing overall patient outcomes. The exploration of CSC signaling pathways represents a key step towards a brighter future in the battle against cancer.
Collapse
Affiliation(s)
- Saptarshi Sinha
- National Institute of Biomedical Genomics, Kalyani, West Bengal, India
| | | | - Subhajit Chatterjee
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, MD, United States.
| |
Collapse
|
10
|
Saad HA, Baz A, Riad M, Eraky ME, El-Taher A, Farid MI, Sharaf K, Said HEM, Ibrahim LA. Tumor microenvironment and immune system preservation in early-stage breast cancer: routes for early recurrence after mastectomy and treatment for lobular and ductal forms of disease. BMC Immunol 2024; 25:9. [PMID: 38273260 PMCID: PMC10809557 DOI: 10.1186/s12865-023-00591-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 12/07/2023] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND Intra-ductal cancer (IDC) is the most common type of breast cancer, with intra-lobular cancer (ILC) coming in second. Surgery is the primary treatment for early stage breast cancer. There are now irrefutable data demonstrating that the immune context of breast tumors can influence growth and metastasis. Adjuvant chemotherapy may be administered in patients who are at a high risk of recurrence. Our goal was to identify the processes underlying both types of early local recurrences. METHODS This was a case-control observational study. Within 2 years of receiving adjuvant taxan and anthracycline-based chemotherapy, as well as modified radical mastectomy (MRM), early stage IDC and ILC recurred. Vimentin, α-smooth muscle actin (SMA), platelet-derived growth factor (PDGF), matrix metalloproteinase (MMP1), and clustered differentiation (CD95) were investigated. RESULTS Of the samples in the ductal type group, 25 showed local recurrence, and 25 did not. Six individuals in the lobular-type group did not experience recurrence, whereas seven did. Vimentin (p = 0.000 and 0.021), PDGF (p = 0.000 and 0.002), and CD95 (p = 0.000 and 0.045) expressions were significantly different in ductal and lobular carcinoma types, respectively. Measurement of ductal type was the sole significant difference found in MMP1 (p = 0.000) and α-SMA (p = 0.000). α-SMA and CD95 were two variables that helped the recurrence mechanism in the ductal type according to the pathway analysis. In contrast, the CD95 route is a recurrent mechanism for the lobular form. CONCLUSIONS While the immune system plays a larger role in ILC, the tumor microenvironment and immune system both influence the recurrence of IDC. According to this study, improving the immune system may be a viable cancer treatment option.
Collapse
Affiliation(s)
- Hassan A Saad
- Surgical Department, Faculty of Medicine, Zagazig University, Zagazig City, 44661, Egypt.
| | - Azza Baz
- Surgical Department, Alahrar Teaching Hospital, Zagazig University, Zagazig City, 55971, Egypt
| | - Mohamed Riad
- Surgical Department, Faculty of Medicine, Zagazig University, Zagazig City, 44661, Egypt
| | - Mohamed E Eraky
- Surgical Department, Faculty of Medicine, Zagazig University, Zagazig City, 44661, Egypt
| | - Ahmed El-Taher
- Surgical Department, Faculty of Medicine, Zagazig University, Zagazig City, 44661, Egypt
| | - Mohamed I Farid
- Surgical Department, Faculty of Medicine, Zagazig University, Zagazig City, 44661, Egypt
| | - Khaled Sharaf
- Surgical Department, Faculty of Medicine, Zagazig University, Zagazig City, 44661, Egypt
| | - Huda E M Said
- Clinical Pathology Department, Faculty of Medicine, Zagazig University, Zagazig City, 55971, Egypt
| | - Lotfy A Ibrahim
- Surgical Department, AlAzhar University, Nasr City, Cairo, 55888, Egypt
| |
Collapse
|
11
|
Luo F, Wang X, Ye C, Sun H. Microbial Biomarkers in Liquid Biopsy for Cancer: An Overview and Future Directions. Cancer Control 2024; 31:10732748241292019. [PMID: 39431347 PMCID: PMC11500238 DOI: 10.1177/10732748241292019] [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: 04/20/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 10/22/2024] Open
Abstract
In recent years, the relationship between microbes and tumors has led to a new wave of scholarly pursuits. Due to the growing awareness of the importance of microbiota, including those within tumors, for cancer onset, progression, metastasis, and treatment, researchers have come to understand that microbiota and the tumor microenvironment together form a dynamic and complex ecosystem. Liquid biopsy technology, a non-invasive and easily repeatable method for sample collection, combined with emerging multi-omics techniques, allows for a more comprehensive and in-depth exploration of microbial signals and characteristics in bodily fluids. Microbial biomarkers hold immense potential in the early diagnosis, treatment stratification, and prognosis prediction of cancer. In this review, we describe the significant potential of microbial biomarkers in liquid biopsy for clinical applications in cancer, including early diagnosis, predicting treatment responses, and prognosis. Moreover, we discuss current limitations and potential solutions related to microbial biomarkers. This review aims to provide an overview and future directions of microbial biomarkers in liquid biopsy for cancer clinical practice.
Collapse
Affiliation(s)
| | - Xinyue Wang
- Xinyue Wang, MB, Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, 253# Gongye Road, Guangzhou 510280, China.
| | | | - Haitao Sun
- Xinyue Wang, MB, Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, The Second School of Clinical Medicine, Southern Medical University, 253# Gongye Road, Guangzhou 510280, China.
| |
Collapse
|
12
|
Zhou Y, Ye Z, Wei W, Zhang M, Huang F, Li J, Cai C. Macrophages maintain mammary stem cell activity and mammary homeostasis via TNF-α-PI3K-Cdk1/Cyclin B1 axis. NPJ Regen Med 2023; 8:23. [PMID: 37130846 PMCID: PMC10154328 DOI: 10.1038/s41536-023-00296-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 04/20/2023] [Indexed: 05/04/2023] Open
Abstract
Adult stem cell niche is a special environment composed of a variety stromal cells and signals, which cooperatively regulate tissue development and homeostasis. It is of great interest to study the role of immune cells in niche. Here, we show that mammary resident macrophages regulate mammary epithelium cell division and mammary development through TNF-α-Cdk1/Cyclin B1 axis. In vivo, depletion of macrophages reduces the number of mammary basal cells and mammary stem cells (MaSCs), while increases mammary luminal cells. In vitro, we establish a three-dimensional culture system in which mammary basal cells are co-cultured with macrophages, and interestingly, macrophage co-culture promotes the formation of branched functional mammary organoids. Moreover, TNF-α produced by macrophages activates the intracellular PI3K/Cdk1/Cyclin B1 signaling in mammary cells, thereby maintaining the activity of MaSCs and the formation of mammary organoids. Together, these findings reveal the functional significance of macrophageal niche and intracellular PI3K/Cdk1/Cyclin B1 axis for maintaining MaSC activity and mammary homeostasis.
Collapse
Affiliation(s)
- Yu Zhou
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Zi Ye
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Wei Wei
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Mengna Zhang
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Fujing Huang
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Jinpeng Li
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China.
| | - Cheguo Cai
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China.
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
| |
Collapse
|
13
|
Xiao Z, Wang X, Chen X, Zhou J, Zhu H, Zhang J, Deng W. Prognostic role of preoperative inflammatory markers in postoperative patients with colorectal cancer. Front Oncol 2023; 13:1064343. [PMID: 37064153 PMCID: PMC10095152 DOI: 10.3389/fonc.2023.1064343] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Background Inflammatory response markers are prognostic factors for several cancers, but their role in postoperative colorectal cancer (CRC) is unclear. The purpose was to evaluate the role of preoperative Neutrophil-to-Lymphocyte ratio (NLR), Platelet-to-Lymphocyte-ratio (PLR), and Lymphocyte-to-Monocyte ratio (LMR) in the prognosis of postoperative CRC patients. Methods We retrospectively reviewed 448 CRC patients who had undergone surgical resection from December 2015 to December 2017 in our hospital. The plasma NLR, PLR, LMR, CEA, and CA19-9 were collected within 2 weeks before the operation. We recorded the clinical characteristics and survival data by reviewing medical records and phone calls. We analyzed preoperative inflammatory markers and clinical features using Pearson chi-squared tests or Fisher's tests. Uni- and multivariate Cox regression analyses were performed, and overall survival (OS) was estimated with the Kaplan-Meier method. Results High NLR and PLR were associated with worse overall survival in postoperative CRC (HR = 2.140, 95%CI = (1.488-3.078), P < 0.001; HR =1.820, 95%CI = (1.271-2.605), P = 0.001). High LMR was associated with improved overall survival in postoperative CRC (HR = 0.341, 95%CI = (0.188-0.618), P < 0.001). In the multivariate regression analysis, the increase of NLR resulted in an increase in the risk of death (HR = 1.678, 95%CI = (1.114-2.527), P = 0.013), and for the LMR, a reduction of the risk of death (HR = 0.480, 95%CI = (0.256 - 0.902), P = 0.023). Moreover, TNM stage, CA-199, CEA, nerve or vascular invasion (NVI) and adjuvant chemotherapy after surgery also were associated with worse overall survival in postoperative CRC. Conclusion Current evidence indicates that preoperative inflammatory markers NLR, LMR, and PLR are associated with overall survival in postoperative patients with colorectal cancer. NLR is an independent risk factor, and LMR is an independent protective factor in CRC patients after surgery.
Collapse
Affiliation(s)
- Zilong Xiao
- Department of General Surgery, First Affiliated Hospital of Nanchang University, Nanchang, China
- Laboratory of Digestive Surgery, Nanchang University, Nanchang, China
| | - Xinxin Wang
- Department of Radiation Oncology, The Third Hospital of Nanchang, Nanchang, China
| | - Xiaoxiao Chen
- Department of General Surgery, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jiawei Zhou
- Department of General Surgery, First Affiliated Hospital of Nanchang University, Nanchang, China
- Laboratory of Digestive Surgery, Nanchang University, Nanchang, China
| | - Haitao Zhu
- Department of Intensive Care Unit, Jining Public Health Medical Center, Jining, China
| | - Jiangnan Zhang
- Department of General Surgery, First Affiliated Hospital of Nanchang University, Nanchang, China
- Laboratory of Digestive Surgery, Nanchang University, Nanchang, China
| | - Wensheng Deng
- Department of General Surgery, First Affiliated Hospital of Nanchang University, Nanchang, China
- Laboratory of Digestive Surgery, Nanchang University, Nanchang, China
| |
Collapse
|
14
|
Wang X, Bai Y, Zhang F, Li D, Chen K, Wu R, Tang Y, Wei X, Han P. Prognostic value of COL10A1 and its correlation with tumor-infiltrating immune cells in urothelial bladder cancer: A comprehensive study based on bioinformatics and clinical analysis validation. Front Immunol 2023; 14:955949. [PMID: 37006317 PMCID: PMC10063846 DOI: 10.3389/fimmu.2023.955949] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 02/22/2023] [Indexed: 03/19/2023] Open
Abstract
IntroductionBladder cancer (BLCA) is one of the most lethal diseases. COL10A1 is secreted small-chain collagen in the extracellular matrix associated with various tumors, including gastric, colon, breast, and lung cancer. However, the role of COL10A1 in BLCA remains unclear. This is the first research focusing on the prognostic value of COL10A1 in BLCA. In this research, we aimed to uncover the association between COL10A1 and the prognosis, as well as other clinicopathological parameters in BLCA.MethodsWe obtained gene expression profiles of BLCA and normal tissues from the TCGA, GEO, and ArrayExpress databases. Immunohistochemistry staining was performed to investigate the protein expression and prognostic value of COL10A1 in BLCA patients. GO and KEGG enrichment along with GSEA analyses were performed to reveal the biological functions and potential regulatory mechanisms of COL10A1 based on the gene co-expression network. We used the “maftools” R package to display the mutation profiles between the high and low COL10A1 groups. GIPIA2, TIMER, and CIBERSORT algorithms were utilized to explore the effect of COL10A1 on the tumor immune microenvironment.ResultsWe found that COL10A1 was upregulated in the BLCA samples, and increased COL10A1 expression was related to poor overall survival. Functional annotation of 200 co-expressed genes positively correlated with COL10A1 expression, including GO, KEGG, and GSEA enrichment analyses, indicated that COL10A1 was basically involved in the extracellular matrix, protein modification, molecular binding, ECM-receptor interaction, protein digestion and absorption, focal adhesion, and PI3K-Akt signaling pathway. The most commonly mutated genes of BLCA were different between high and low COL10A1 groups. Tumor immune infiltrating analyses showed that COL10A1 might have an essential role in recruiting infiltrating immune cells and regulating immunity in BLCA, thus affecting prognosis. Finally, external datasets and biospecimens were used, and the results further validated the aberrant expression of COL10A1 in BLCA samples.ConclusionsIn conclusion, our study demonstrates that COL10A1 is an underlying prognostic and predictive biomarker in BLCA.
Collapse
|
15
|
Yang Q, Kennicott K, Zhu R, Kim J, Wakefield H, Studener K, Liang Y. Sex hormone influence on female-biased autoimmune diseases hints at puberty as an important factor in pathogenesis. Front Pediatr 2023; 11:1051624. [PMID: 36793337 PMCID: PMC9923181 DOI: 10.3389/fped.2023.1051624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 01/10/2023] [Indexed: 01/31/2023] Open
Abstract
The majority of autoimmune diseases affect more women than men, suggesting an important role for sex hormones in regulating immune response. Current research supports this idea, highlighting the importance of sex hormones in both immune and metabolic regulation. Puberty is characterized by drastic changes in sex hormone levels and metabolism. These pubertal changes may be what forms the gulf between men and women in sex bias towards autoimmunity. In this review, a current perspective on pubertal immunometabolic changes and their impact on the pathogenesis of a select group of autoimmune diseases is presented. SLE, RA, JIA, SS, and ATD were focused on in this review for their notable sex bias and prevalence. Due to both the scarcity of pubertal autoimmune data and the differences in mechanism or age-of-onset in juvenile analogues often beginning prior to pubertal changes, data on the connection between the specific adult autoimmune diseases and puberty often relies on sex hormone influence in pathogenesis and established sex differences in immunity that begin during puberty.
Collapse
Affiliation(s)
- Qianfan Yang
- School of Medicine and Public Health, Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, United
| | - Kameron Kennicott
- Departments of Physiology and Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
| | - Runqi Zhu
- Departments of Physiology and Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
| | - Jooyong Kim
- School of Medicine and Public Health, Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, United
| | - Hunter Wakefield
- School of Medicine and Public Health, Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, United
| | - Katelyn Studener
- School of Medicine and Public Health, Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, United
| | - Yun Liang
- Departments of Physiology and Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
- Correspondence: Yun Liang
| |
Collapse
|
16
|
Breast cancer prevention by short-term inhibition of TGFβ signaling. Nat Commun 2022; 13:7558. [PMID: 36476730 PMCID: PMC9729304 DOI: 10.1038/s41467-022-35043-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 11/16/2022] [Indexed: 12/12/2022] Open
Abstract
Cancer prevention has a profound impact on cancer-associated mortality and morbidity. We previously identified TGFβ signaling as a candidate regulator of mammary epithelial cells associated with breast cancer risk. Here, we show that short-term TGFBR inhibitor (TGFBRi) treatment of peripubertal ACI inbred and Sprague Dawley outbred rats induces lasting changes and prevents estrogen- and carcinogen-induced mammary tumors, respectively. We identify TGFBRi-responsive cell populations by single cell RNA-sequencing, including a unique epithelial subpopulation designated secretory basal cells (SBCs) with progenitor features. We detect SBCs in normal human breast tissues and find them to be associated with breast cancer risk. Interactome analysis identifies SBCs as the most interactive cell population and the main source of insulin-IGF signaling. Accordingly, inhibition of TGFBR and IGF1R decrease proliferation of organoid cultures. Our results reveal a critical role for TGFβ in regulating mammary epithelial cells relevant to breast cancer and serve as a proof-of-principle cancer prevention strategy.
Collapse
|
17
|
Feng TY, Azar FN, Dreger SA, Buchta Rosean C, McGinty MT, Putelo AM, Kolli SH, Carey MA, Greenfield S, Fowler WJ, Robinson SD, Rutkowski MR. Reciprocal Interactions Between the Gut Microbiome and Mammary Tissue Mast Cells Promote Metastatic Dissemination of HR+ Breast Tumors. Cancer Immunol Res 2022; 10:1309-1325. [PMID: 36040846 PMCID: PMC9633553 DOI: 10.1158/2326-6066.cir-21-1120] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 06/02/2022] [Accepted: 08/25/2022] [Indexed: 11/16/2022]
Abstract
Establishing commensal dysbiosis, defined as an inflammatory gut microbiome with low biodiversity, before breast tumor initiation, enhances early dissemination of hormone receptor-positive (HR+) mammary tumor cells. Here, we sought to determine whether cellular changes occurring in normal mammary tissues, before tumor initiation and in response to dysbiosis, enhanced dissemination of HR+ tumors. Commensal dysbiosis increased both the frequency and profibrogenicity of mast cells in normal, non-tumor-bearing mammary tissues, a phenotypic change that persisted after tumor implantation. Pharmacological and adoptive transfer approaches demonstrated that profibrogenic mammary tissue mast cells from dysbiotic animals were sufficient to enhance dissemination of HR+ tumor cells. Using archival HR+ patient samples, we determined that enhanced collagen levels in tumor-adjacent mammary tissue positively correlated with mast cell abundance and HR+ breast cancer recurrence. Together, these data demonstrate that mast cells programmed by commensal dysbiosis activate mammary tissue fibroblasts and orchestrate early dissemination of HR+ breast tumors.
Collapse
Affiliation(s)
- Tzu-Yu Feng
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, VA, USA
| | - Francesca N. Azar
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, VA, USA
| | - Sally A. Dreger
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Claire Buchta Rosean
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, VA, USA
| | - Mitchell T. McGinty
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, VA, USA
| | - Audrey M. Putelo
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, VA, USA
| | - Sree H. Kolli
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, VA, USA
| | - Maureen A. Carey
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, VA, USA
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, VA, USA
| | - Stephanie Greenfield
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, VA, USA
| | - Wesley J. Fowler
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Stephen D. Robinson
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, UK
| | - Melanie R. Rutkowski
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, VA, USA
| |
Collapse
|
18
|
Jungles KM, Holcomb EA, Pearson AN, Jungles KR, Bishop CR, Pierce LJ, Green MD, Speers CW. Updates in combined approaches of radiotherapy and immune checkpoint inhibitors for the treatment of breast cancer. Front Oncol 2022; 12:1022542. [PMID: 36387071 PMCID: PMC9643771 DOI: 10.3389/fonc.2022.1022542] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 09/27/2022] [Indexed: 12/05/2022] Open
Abstract
Breast cancer is the most prevalent non-skin cancer diagnosed in females and developing novel therapeutic strategies to improve patient outcomes is crucial. The immune system plays an integral role in the body’s response to breast cancer and modulating this immune response through immunotherapy is a promising therapeutic option. Although immune checkpoint inhibitors were recently approved for the treatment of breast cancer patients, not all patients respond to immune checkpoint inhibitors as a monotherapy, highlighting the need to better understand the biology underlying patient response. Additionally, as radiotherapy is a critical component of breast cancer treatment, understanding the interplay of radiation and immune checkpoint inhibitors will be vital as recent studies suggest that combined therapies may induce synergistic effects in preclinical models of breast cancer. This review will discuss the mechanisms supporting combined approaches with radiotherapy and immune checkpoint inhibitors for the treatment of breast cancer. Moreover, this review will analyze the current clinical trials examining combined approaches of radiotherapy, immunotherapy, chemotherapy, and targeted therapy. Finally, this review will evaluate data regarding treatment tolerance and potential biomarkers for these emerging therapies aimed at improving breast cancer outcomes.
Collapse
Affiliation(s)
- Kassidy M. Jungles
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, United States
| | - Erin A. Holcomb
- Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Ashley N. Pearson
- Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Kalli R. Jungles
- Department of Biology, Saint Mary’s College, Notre Dame, IN, United States
| | - Caroline R. Bishop
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, United States
| | - Lori J. Pierce
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
| | - Michael D. Green
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, United States
- Department of Radiation Oncology, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI, United States
- *Correspondence: Michael D. Green, ; Corey W. Speers,
| | - Corey W. Speers
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
- Department of Radiation Oncology, University Hospitals Cleveland Medical Center, Case Western Reserve University, Case Comprehensive Cancer Center, Cleveland, OH, United States
- *Correspondence: Michael D. Green, ; Corey W. Speers,
| |
Collapse
|
19
|
Zhou W, Li Y, Gu D, Xu J, Wang R, Wang H, Liu C. High expression COL10A1 promotes breast cancer progression and predicts poor prognosis. Heliyon 2022; 8:e11083. [PMID: 36281404 PMCID: PMC9586897 DOI: 10.1016/j.heliyon.2022.e11083] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/28/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022] Open
Abstract
Background As a common malignant disease in females, breast cancer (BCa) causes increasing numbers of cancer-related death. Collagen X alpha 1 chain (COL10A1) plays a critical role in the oncogenesis and progression of malignant tumors. However, a systematic analysis of COL10A1 in BCa has not been conducted. Methods The COL10A1 expression level and prognostic value in BCa were defined through the Cancer Genome Atlas (TCGA) as well as the Kaplan-Meier plotter data respectively. The expression pattern of COL10A1 was subsequently confirmed on tissue microarray (TMA) by immunochemistry (IHC) staining. Moreover, cellular functional assays which aimed to evaluate cell proliferation, migration, invasion, and apoptosis, were conducted to investigate the oncogenic activity of COL10A1 in BCa. Then, Tumor Immune Estimation Resource (TIMER) was adopted to determine the association between COL10A1 expression and immune cell infiltration. Results Bioinformatics analysis revealed that COL10A1 was significantly overexpressed and had notable prognostic value, especially for distant metastasis-free survival (DMFS) in BCa. Moreover, IHC analysis of 140 BCa tissues on TMA chips exhibited the overexpression of COL10A1 was correlated to advanced clinical stage, poor overall survival (OS), and worse recurrence-free survival (RFS). Besides, knockdown of COL10A1 remarkably suppressed cell proliferation, migration, and invasion in BCa cells, and notably promoted cell apoptosis as well. Furthermore, COL10A1 was positively associated with immune cell infiltration including B cell, CD8+ T cell, CD4+ T cell, macrophage, neutrophil, and dendritic cell. Conclusion The results revealed that COL10A1 is a novel oncogene and could serve as a potential prognostic biomarker in BCa. Besides, the downregulation of COL10A1 could inhibit BCa progression, which could be a potential target for BCa therapy.
Collapse
|
20
|
Al-Khaldi S, Almohanna F, Barnawi R, Fallatah M, Islam SS, Ghebeh H, Al-Alwan M. Fascin is essential for mammary gland lactogenesis. Dev Biol 2022; 492:25-36. [PMID: 36152869 DOI: 10.1016/j.ydbio.2022.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 05/29/2022] [Accepted: 09/16/2022] [Indexed: 11/03/2022]
Abstract
Fascin expression has commonly been observed in certain subtypes of breast cancer, where its expression is associated with poor clinical outcome. However, its role in normal mammary gland development has not been elucidated. Here, we used a fascin knockout mouse model to assess its role in normal mammary gland morphogenesis and lactation. Fascin knockout was not embryonically lethal, and its effect on the litter size or condition at birth was minimal. However, litter survival until the weaning stage significantly depended on fascin expression solely in the nursing dams. Accordingly, pups that nursed from fascin-/- dams had smaller milk spots in their abdomen, suggesting a lactation defect in the nursing dams. Mammary gland whole-mounts of pregnant and lactating fascin-/- mice showed significantly reduced side branching and alveologenesis. Despite a typical composition of basal, luminal, and stromal subsets of mammary cells and normal ductal architecture of myoepithelial and luminal layers, the percentage of alveolar progenitors (ALDH+) in fascin-/- epithelial fraction was significantly reduced. Further in-depth analyses of fascin-/- mammary glands showed a significant reduction in the expression of Elf5, the master regulator of alveologenesis, and a decrease in the activity of its downstream target p-STAT5. In agreement, there was a significant reduction in the expression of the milk proteins, whey acidic protein (WAP), and β-casein in fascin-/- mammary glands. Collectively, our data demonstrate, for the first time, the physiological role of fascin in normal mammary gland lactogenesis, an addition that could reveal its contribution to breast cancer initiation and progression.
Collapse
Affiliation(s)
- Samiyah Al-Khaldi
- National Center for Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Sciences and Technology, Riyadh, Saudi Arabia.
| | | | | | - Mohannad Fallatah
- National Center for Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Sciences and Technology, Riyadh, Saudi Arabia.
| | - Syed S Islam
- Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia; Collage of Medicine, Al-Faisal University, Riyadh, Saudi Arabia.
| | - Hazem Ghebeh
- Stem Cell and Tissue Re-Engineering Program, Saudi Arabia; Collage of Medicine, Al-Faisal University, Riyadh, Saudi Arabia.
| | - Monther Al-Alwan
- Stem Cell and Tissue Re-Engineering Program, Saudi Arabia; Collage of Medicine, Al-Faisal University, Riyadh, Saudi Arabia.
| |
Collapse
|
21
|
Tran T, Lavillegrand JR, Lereverend C, Esposito B, Cartier L, Montabord M, Tran-Rajau J, Diedisheim M, Gruel N, Ouguerram K, Paolini L, Lenoir O, Pinteaux E, Brabencova E, Tanchot C, Urquia P, Lehmann-Che J, Le Naour R, Merrouche Y, Stockmann C, Mallat Z, Tedgui A, Ait-Oufella H, Tartour E, Potteaux S. Mild dyslipidemia accelerates tumorigenesis through expansion of Ly6C hi monocytes and differentiation to pro-angiogenic myeloid cells. Nat Commun 2022; 13:5399. [PMID: 36104342 PMCID: PMC9475043 DOI: 10.1038/s41467-022-33034-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 08/24/2022] [Indexed: 11/08/2022] Open
Abstract
Cancer and cardiovascular disease (CVD) share common risk factors such as dyslipidemia, obesity and inflammation. However, the role of pro-atherogenic environment and its associated low-grade inflammation in tumor progression remains underexplored. Here we show that feeding C57BL/6J mice with a non-obesogenic high fat high cholesterol diet (HFHCD) for two weeks to induce mild dyslipidemia, increases the pool of circulating Ly6Chi monocytes available for initial melanoma development, in an IL-1β-dependent manner. Descendants of circulating myeloid cells, which accumulate in the tumor microenvironment of mice under HFHCD, heighten pro-angiogenic and immunosuppressive activities locally. Limiting myeloid cell accumulation or targeting VEGF-A production by myeloid cells decrease HFHCD-induced tumor growth acceleration. Reverting the HFHCD to a chow diet at the time of tumor implantation protects against tumor growth. Together, these data shed light on cross-disease communication between cardiovascular pathologies and cancer.
Collapse
Affiliation(s)
- Thi Tran
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
| | | | - Cedric Lereverend
- Université de Reims Champagne Ardenne, IRMAIC EA 7509, 51097, Reims, France
| | - Bruno Esposito
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
| | - Lucille Cartier
- Université de Reims Champagne Ardenne, IRMAIC EA 7509, 51097, Reims, France
- Département de Recherche, Institut Godinot, 51100, Reims, France
| | | | | | - Marc Diedisheim
- Service de diabétologie, Hôpital Cochin APHP. GlandOmics, Cheverny, Paris, France
| | - Nadège Gruel
- INSERM U830, Équipe Labellisée LNCC, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, Institut Curie Research Centre, Institut Curie, 75005, Paris, France
- Department of Translational Research, Institut Curie Research Centre, Institut Curie, 75005, Paris, France
| | | | - Lea Paolini
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
| | - Olivia Lenoir
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
| | - Emmanuel Pinteaux
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Eva Brabencova
- Département de Recherche, Institut Godinot, 51100, Reims, France
| | - Corinne Tanchot
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
| | - Pauline Urquia
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
| | - Jacqueline Lehmann-Che
- Université Paris Cité, INSERM, U976 HIPI, F-75010, Paris, France
- Molecular Oncology Unit, Saint Louis Hospital, APHP, F-75010, Paris, France
| | - Richard Le Naour
- Université de Reims Champagne Ardenne, IRMAIC EA 7509, 51097, Reims, France
| | - Yacine Merrouche
- Université de Reims Champagne Ardenne, IRMAIC EA 7509, 51097, Reims, France
- Département de Recherche, Institut Godinot, 51100, Reims, France
| | - Christian Stockmann
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
- University of Zurich, Institute of Anatomy, Zurich, Switzerland
- Comprehensive Cancer Center Zurich, Zurich, Switzerland
| | - Ziad Mallat
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Alain Tedgui
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
| | | | - Eric Tartour
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
- AP-HP Hôpital Européen Georges Pompidou. Service d'immunologie, Paris, France
| | - Stephane Potteaux
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France.
- Université Paris Cité, INSERM, U976 HIPI, F-75010, Paris, France.
| |
Collapse
|
22
|
Qian W, Zhang Y, Long Y, Yang W, Hu R, Li J, Leng Y, Liu X, Li QX, Wan X, Wei X. Probiotic Lactobacillus brevis CLB3 prevents azoxymethane/dextran sulfate sodium induced colon carcinogenesis in mice by reducing amino acid transport and IL-17A levels and repressing the IL-6/AKT/p-STAT3 signaling pathway. FOOD QUALITY AND SAFETY 2022. [DOI: 10.1093/fqsafe/fyac038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Abstract
Amino acid intake plays a crucial role in the Warburg effect of cancer. The gut microbes could regulate intestinal amino acid metabolism. However, it is still unknown whether a probiotic therapy can protect the host from intestinal tumor invasion by reducing amino acid intake. With in vitro methods, three acid tolerant strains from fermented pickles were screened out. Using AOM/DSS induced colon cancer models, we evaluated the therapeutic effects of Lactobacillus brevis CLB3, Lactobacillus plantarum XLP and Lactobacillus johnsonii CM on model mice. Their functional mechanism were further explained through anatomy section, qRT-PCR, Western blot, and immunohistochemical staining analyses as well as database mining and gut culturomics. The L. brevis CLB3 treatment significantly improved clinical signs and symptoms of colon cancer, alleviated colon damage, and inhibited colon carcinogenesis in mice. In addition, this treatment significantly increased gut cultivable Lactobacillus abundance, inhibited the expression and translation levels of the tumor metabolism-related solute carrier (SLC) amino acid transporter including SLC7A5 and SLC7A11, lowered circulating IL-6 and IL-17A levels, and improved the accumulation of tumor-infiltrating lymphocytes and cancer proliferation factors. These findings suggest that L. brevis CLB3 can reduce amino acid transport, inhibit mTOR signaling and enhance intestinal anti-tumor immune responses, which provides a potential targeting amino acid transporters strategy for preventing colorectal cancer.
Collapse
|
23
|
Hitchcock J, Hughes K, Pensa S, Lloyd-Lewis B, Watson CJ. The immune environment of the mammary gland fluctuates during post-lactational regression and correlates with tumour growth rate. Development 2022; 149:275060. [PMID: 35420674 PMCID: PMC9124574 DOI: 10.1242/dev.200162] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 04/04/2022] [Indexed: 01/02/2023]
Abstract
Post-lactational mammary gland regression encompasses extensive programmed cell death and removal of milk-producing epithelial cells, breakdown of extracellular matrix components and redifferentiation of stromal adipocytes. This highly regulated involution process is associated with a transient increased risk of breast cancer in women. Using a syngeneic tumour model, we show that tumour growth is significantly altered depending on the stage of involution at which tumour cells are implanted. Tumour cells injected at day 3 involution grew faster than those in nulliparous mice, whereas tumours initiated at day 6 involution grew significantly slower. These differences in tumour progression correlate with distinct changes in innate immune cells, in particular among F4/80-expressing macrophages and among TCRδ+ unconventional T cells. Breast cancer post-pregnancy risk is exacerbated in older first-time mothers and, in our model, initial tumour growth is moderately faster in aged mice compared with young mice. Our results have implications for breast cancer risk and the use of anti-inflammatory therapeutics for postpartum breast cancers. Summary: Mammary gland involution is associated with dynamic changes in immune cell types and numbers at different stages that correlates with the initial rate of growth of implanted tumour cells.
Collapse
Affiliation(s)
- Jessica Hitchcock
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| | - Katherine Hughes
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, UK
| | - Sara Pensa
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK
| | - Bethan Lloyd-Lewis
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
- School of Cellular and Molecular Medicine, University of Bristol, University Walk, Bristol BS8 1TD, UK
| | - Christine J. Watson
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| |
Collapse
|
24
|
Gao Z, Mi R, Cheng Z, Li X, Zeng H, Wu G, Zhao J, Zhang W, Ye J. Integrated Metabolomics and Network Pharmacology Revealed Hong-Hua-Xiao-Yao Tablet's Effect of Mediating Hormone Synthesis in the Treatment of Mammary Gland Hyperplasia. Front Pharmacol 2022; 13:788019. [PMID: 35177987 PMCID: PMC8846323 DOI: 10.3389/fphar.2022.788019] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 01/07/2022] [Indexed: 11/13/2022] Open
Abstract
Hong-Hua-Xiao-Yao Tablet (HHXYT) is a traditional Chinese medicine (TCM) formula that has been approved for the treatment of mammary gland hyperplasia (MGH), but its mechanism of action is unclear. In this study, a strategy that integrated metabolomics and network pharmacology was applied to systemically reveal the mechanism of HHXYT in the treatment of MGH. Our pharmacodynamic study indicated that the proliferation of mammary gland was inhibited in rats, and serum-level disorder of estradiol and progesterone was reversed after HHXYT treatment. 54 compounds absorbed in rat plasma were identified after administration of HHXYT. The serum metabolome revealed 58 endogenous differential metabolites, of which 31% were steroid lipids metabolites, with steroid hormone biosynthesis being the most significant metabolic module. 7 targets, 6 herbs, and 17 ingredients were found to play key roles in HHXYT’s treatment of MGH. 3 of the 7 key targets (CYP11A1, HSD3B2, and CYP17A1) were directly involved in androgen synthesis, while 2 targets (AR and ESR1) were receptors for the direct action of androgens and estrogens. Molecular docking was utilized to confirm the bindings between the 5 targets and their corresponding compounds. In an in vitro test, HHXYT (50 µg/ml) and its ingredient formononetin (3.2, 6.3, and 12.5 µM) were found to significantly reduce the increase of testosterone level induced by dexamethasone (10 µM) in thecal cells. In summary, this study illustrated that the mechanism of HHXYT’s treatment of MGH was to regulate hormone disorder. HHXYT could reduce estrogen-stimulated hyperplasia by inhibiting the production of its precursor androgen.
Collapse
Affiliation(s)
- Ziqing Gao
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Rui Mi
- School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Zhaoxi Cheng
- School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Xiaofeng Li
- School of Pharmacy, Fudan University, Shanghai, China
| | - Huawu Zeng
- School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Gaosong Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing Zhao
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weidong Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Ji Ye
- School of Pharmacy, Second Military Medical University, Shanghai, China
| |
Collapse
|
25
|
Korte BG, Giese MA, Ramakrishnan G, Ma S, Bennin D, Rindy J, Dewey CN, Huttenlocher A. Cell Type-Specific Transcriptome Profiling Reveals a Role for Thioredoxin During Tumor Initiation. Front Immunol 2022; 13:818893. [PMID: 35250998 PMCID: PMC8891495 DOI: 10.3389/fimmu.2022.818893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 01/25/2022] [Indexed: 01/27/2023] Open
Abstract
Neutrophils in the tumor microenvironment exhibit altered functions. However, the changes in neutrophil behavior during tumor initiation remain unclear. Here we used Translating Ribosomal Affinity Purification (TRAP) and RNA sequencing to identify neutrophil, macrophage and transformed epithelial cell transcriptional changes induced by oncogenic RasG12V in larval zebrafish. We found that transformed epithelial cells and neutrophils, but not macrophages, had significant changes in gene expression in larval zebrafish. Interestingly, neutrophils had more significantly down-regulated genes, whereas gene expression was primarily upregulated in transformed epithelial cells. The antioxidant, thioredoxin (txn), a small thiol that regulates reduction-oxidation (redox) balance, was upregulated in transformed keratinocytes and neutrophils in response to oncogenic Ras. To determine the role of thioredoxin during tumor initiation, we generated a zebrafish thioredoxin mutant. We observed an increase in wound-induced reactive oxygen species signaling and neutrophil recruitment in thioredoxin-deficient zebrafish. Transformed keratinocytes also showed increased proliferation and reduced apoptosis in thioredoxin-deficient larvae. Using live imaging, we visualized neutrophil behavior near transformed cells and found increased neutrophil recruitment and altered motility dynamics. Finally, in the absence of neutrophils, transformed keratinocytes no longer exhibited increased proliferation in thioredoxin mutants. Taken together, our findings demonstrate that tumor initiation induces changes in neutrophil gene expression and behavior that can impact proliferation of transformed cells in the early tumor microenvironment.
Collapse
Affiliation(s)
- Benjamin G. Korte
- Department of Pediatrics and Medical Microbiology and Immunology, University of Wisconsin – Madison, Madison, WI, United States
- Cancer Biology Graduate Program, University of Wisconsin – Madison, Madison, WI, United States
| | - Morgan A. Giese
- Department of Pediatrics and Medical Microbiology and Immunology, University of Wisconsin – Madison, Madison, WI, United States
- Cellular and Molecular Biology Graduate Program, University of Wisconsin – Madison, Madison, WI, United States
| | - Gayathri Ramakrishnan
- Department of Pediatrics and Medical Microbiology and Immunology, University of Wisconsin – Madison, Madison, WI, United States
- Cancer Biology Graduate Program, University of Wisconsin – Madison, Madison, WI, United States
| | - Stella Ma
- Department of Pediatrics and Medical Microbiology and Immunology, University of Wisconsin – Madison, Madison, WI, United States
| | - David Bennin
- Department of Pediatrics and Medical Microbiology and Immunology, University of Wisconsin – Madison, Madison, WI, United States
| | - Julie Rindy
- Department of Pediatrics and Medical Microbiology and Immunology, University of Wisconsin – Madison, Madison, WI, United States
| | - Colin N. Dewey
- Department of Biostatistics and Medical Informatics, University of Wisconsin – Madison, Madison, WI, United States
| | - Anna Huttenlocher
- Department of Pediatrics and Medical Microbiology and Immunology, University of Wisconsin – Madison, Madison, WI, United States
- Department of Pediatrics, University of Wisconsin – Madison, Madison, WI, United States
| |
Collapse
|
26
|
Su Y, Santucci-Pereira J, Dang NM, Kanefsky J, Rahulkannan V, Hillegass M, Joshi S, Gurdogan H, Chen Z, Bessonneau V, Rudel R, Ser-Dolansky J, Schneider SS, Russo J. Effects of Pubertal Exposure to Butyl Benzyl Phthalate, Perfluorooctanoic Acid, and Zeranol on Mammary Gland Development and Tumorigenesis in Rats. Int J Mol Sci 2022; 23:ijms23031398. [PMID: 35163327 PMCID: PMC8835802 DOI: 10.3390/ijms23031398] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/18/2022] [Accepted: 01/24/2022] [Indexed: 02/04/2023] Open
Abstract
Endocrine-disrupting chemicals (EDCs)—including butyl benzyl phthalate (BBP), perfluorooctanoic acid (PFOA), and zeranol (α-ZAL, referred to as ZAL hereafter)—can interfere with the endocrine system and produce adverse effects. It remains unclear whether pubertal exposure to low doses of BBP, PFOA, and ZAL has an impact on breast development and tumorigenesis. We exposed female Sprague Dawley rats to BBP, PFOA, or ZAL through gavage for 21 days, starting on day 21, and analyzed their endocrine organs, serum hormones, mammary glands, and transcriptomic profiles of the mammary glands at days 50 and 100. We also conducted a tumorigenesis study for rats treated with PFOA and ZAL using a 7,12-dimethylbenz[a]anthracene (DMBA) model. Our results demonstrated that pubertal exposure to BBP, PFOA, and ZAL affected endocrine organs and serum hormones, and induced phenotypic and transcriptomic changes. The exposure to PFOA + ZAL induced the most phenotypic and transcriptomic changes in the mammary gland. PFOA + ZAL downregulated the expression of genes related to development at day 50, whereas it upregulated genes associated with tumorigenesis at day 100. PFOA + ZAL exposure also decreased rat mammary tumor latency, reduced the overall survival of rats after DMBA challenge, and affected the histopathology of mammary tumors. Therefore, our study suggests that exposure to low doses of EDCs during the pubertal period could induce changes in the endocrine system and mammary gland development in rats. The inhibition of mammary gland development by PFOA + ZAL might increase the risk of developing mammary tumors through activation of signaling pathways associated with tumorigenesis.
Collapse
Affiliation(s)
- Yanrong Su
- The Irma H Russo, MD-Breast Cancer Research Laboratory, Fox Chase Cancer Center-Temple Health, 333 Cottman Avenue, Philadelphia, PA 19111, USA; (J.S.-P.); (N.M.D.); (J.K.); (V.R.); (M.H.); (S.J.); (H.G.); (Z.C.); (J.R.)
- Correspondence:
| | - Julia Santucci-Pereira
- The Irma H Russo, MD-Breast Cancer Research Laboratory, Fox Chase Cancer Center-Temple Health, 333 Cottman Avenue, Philadelphia, PA 19111, USA; (J.S.-P.); (N.M.D.); (J.K.); (V.R.); (M.H.); (S.J.); (H.G.); (Z.C.); (J.R.)
| | - Nhi M. Dang
- The Irma H Russo, MD-Breast Cancer Research Laboratory, Fox Chase Cancer Center-Temple Health, 333 Cottman Avenue, Philadelphia, PA 19111, USA; (J.S.-P.); (N.M.D.); (J.K.); (V.R.); (M.H.); (S.J.); (H.G.); (Z.C.); (J.R.)
| | - Joice Kanefsky
- The Irma H Russo, MD-Breast Cancer Research Laboratory, Fox Chase Cancer Center-Temple Health, 333 Cottman Avenue, Philadelphia, PA 19111, USA; (J.S.-P.); (N.M.D.); (J.K.); (V.R.); (M.H.); (S.J.); (H.G.); (Z.C.); (J.R.)
| | - Vishnuprabha Rahulkannan
- The Irma H Russo, MD-Breast Cancer Research Laboratory, Fox Chase Cancer Center-Temple Health, 333 Cottman Avenue, Philadelphia, PA 19111, USA; (J.S.-P.); (N.M.D.); (J.K.); (V.R.); (M.H.); (S.J.); (H.G.); (Z.C.); (J.R.)
| | - Meardey Hillegass
- The Irma H Russo, MD-Breast Cancer Research Laboratory, Fox Chase Cancer Center-Temple Health, 333 Cottman Avenue, Philadelphia, PA 19111, USA; (J.S.-P.); (N.M.D.); (J.K.); (V.R.); (M.H.); (S.J.); (H.G.); (Z.C.); (J.R.)
| | - Shalina Joshi
- The Irma H Russo, MD-Breast Cancer Research Laboratory, Fox Chase Cancer Center-Temple Health, 333 Cottman Avenue, Philadelphia, PA 19111, USA; (J.S.-P.); (N.M.D.); (J.K.); (V.R.); (M.H.); (S.J.); (H.G.); (Z.C.); (J.R.)
| | - Hafsa Gurdogan
- The Irma H Russo, MD-Breast Cancer Research Laboratory, Fox Chase Cancer Center-Temple Health, 333 Cottman Avenue, Philadelphia, PA 19111, USA; (J.S.-P.); (N.M.D.); (J.K.); (V.R.); (M.H.); (S.J.); (H.G.); (Z.C.); (J.R.)
| | - Zhen Chen
- The Irma H Russo, MD-Breast Cancer Research Laboratory, Fox Chase Cancer Center-Temple Health, 333 Cottman Avenue, Philadelphia, PA 19111, USA; (J.S.-P.); (N.M.D.); (J.K.); (V.R.); (M.H.); (S.J.); (H.G.); (Z.C.); (J.R.)
| | - Vincent Bessonneau
- Silent Spring Institute, 320 Nevada Street, Suite 302, Newton, MA 02460, USA; (V.B.); (R.R.)
| | - Ruthann Rudel
- Silent Spring Institute, 320 Nevada Street, Suite 302, Newton, MA 02460, USA; (V.B.); (R.R.)
| | - Jennifer Ser-Dolansky
- Pioneer Valley Life Sciences Institute, UMASS Chan Medical School-Baystate, Springfield, MA 01199, USA; (J.S.-D.); (S.S.S.)
| | - Sallie S. Schneider
- Pioneer Valley Life Sciences Institute, UMASS Chan Medical School-Baystate, Springfield, MA 01199, USA; (J.S.-D.); (S.S.S.)
| | - Jose Russo
- The Irma H Russo, MD-Breast Cancer Research Laboratory, Fox Chase Cancer Center-Temple Health, 333 Cottman Avenue, Philadelphia, PA 19111, USA; (J.S.-P.); (N.M.D.); (J.K.); (V.R.); (M.H.); (S.J.); (H.G.); (Z.C.); (J.R.)
| |
Collapse
|
27
|
Kozlov AP. Mammalian tumor-like organs. 1. The role of tumor-like normal organs and atypical tumor organs in the evolution of development (carcino-evo-devo). Infect Agent Cancer 2022; 17:2. [PMID: 35012580 PMCID: PMC8751115 DOI: 10.1186/s13027-021-00412-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 12/23/2021] [Indexed: 12/24/2022] Open
Abstract
Background Earlier I hypothesized that hereditary tumors might participate in the evolution of multicellular organisms. I formulated the hypothesis of evolution by tumor neofunctionalization, which suggested that the evolutionary role of hereditary tumors might consist in supplying evolving multicellular organisms with extra cell masses for the expression of evolutionarily novel genes and the origin of new cell types, tissues, and organs. A new theory—the carcino-evo-devo theory—has been developed based on this hypothesis. Main text My lab has confirmed several non-trivial predictions of this theory. Another non-trivial prediction is that evolutionarily new organs if they originated from hereditary tumors or tumor-like structures, should recapitulate some tumor features in their development. This paper reviews the tumor-like features of evolutionarily novel organs. It turns out that evolutionarily new organs such as the eutherian placenta, mammary gland, prostate, the infantile human brain, and hoods of goldfishes indeed have many features of tumors. I suggested calling normal organs, which have many tumor features, the tumor-like organs. Conclusion Tumor-like organs might originate from hereditary atypical tumor organs and represent the part of carcino-evo-devo relationships, i.e., coevolution of normal and neoplastic development. During subsequent evolution, tumor-like organs may lose the features of tumors and the high incidence of cancer and become normal organs without (or with almost no) tumor features.
Collapse
Affiliation(s)
- A P Kozlov
- Vavilov Institute of General Genetics, Russian Academy of Sciences, 3, Gubkina Street, Moscow, Russia, 117971. .,Peter the Great St. Petersburg Polytechnic University, 29, Polytekhnicheskaya Street, St. Petersburg, Russia, 195251.
| |
Collapse
|
28
|
Abstract
Prolactin coordinates with the ovarian steroids to orchestrate mammary development and lactation, culminating in nourishment and an increasingly appreciated array of other benefits for neonates. Its central activities in mammary epithelial growth and differentiation suggest that it plays a role(s) in breast cancer, but it has been challenging to identify its contributions, essential for incorporation into prevention and treatment approaches. Large prospective epidemiologic studies have linked higher prolactin exposure to increased risk, particularly for ER+ breast cancer in postmenopausal women. However, it has been more difficult to determine its actions and clinical consequences in established tumors. Here we review experimental data implicating multiple mechanisms by which prolactin may increase the risk of breast cancer. We then consider the evidence for role(s) of prolactin and its downstream signaling cascades in disease progression and treatment responses, and discuss how new approaches are beginning to illuminate the biology behind the seemingly conflicting epidemiologic and experimental studies of prolactin actions across diverse breast cancers.
Collapse
|
29
|
Hanasoge Somasundara AV, Moss MA, Feigman MJ, Chen C, Cyrill SL, Ciccone MF, Trousdell MC, Vollbrecht M, Li S, Kendall J, Beyaz S, Wilkinson JE, Dos Santos CO. Parity-induced changes to mammary epithelial cells control NKT cell expansion and mammary oncogenesis. Cell Rep 2021; 37:110099. [PMID: 34879282 PMCID: PMC8719356 DOI: 10.1016/j.celrep.2021.110099] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 08/25/2021] [Accepted: 11/15/2021] [Indexed: 12/19/2022] Open
Abstract
Pregnancy reprograms mammary epithelial cells (MECs) to control their responses to pregnancy hormone re-exposure and carcinoma progression. However, the influence of pregnancy on the mammary microenvironment is less clear. Here, we used single-cell RNA sequencing to profile the composition of epithelial and non-epithelial cells in mammary tissue from nulliparous and parous female mice. Our analysis indicates an expansion of γδ natural killer T-like immune cells (NKTs) following pregnancy and upregulation of immune signaling molecules in post-pregnancy MECs. We show that expansion of NKTs following pregnancy is due to elevated expression of the antigen-presenting molecule CD1d on MECs. Loss of CD1d expression on post-pregnancy MECs, or overall lack of activated NKTs, results in mammary oncogenesis. Collectively, our findings illustrate how pregnancy-induced changes modulate the communication between MECs and the immune microenvironment and establish a causal link between pregnancy, the immune microenvironment, and mammary oncogenesis.
Collapse
MESH Headings
- Animals
- Antigens, CD1d/metabolism
- Cell Communication
- Cell Proliferation
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/immunology
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Epithelial Cells/immunology
- Epithelial Cells/metabolism
- Epithelial Cells/pathology
- Female
- Gene Expression Regulation, Neoplastic
- Genes, BRCA1
- Genes, myc
- Lymphocyte Activation
- Mammary Glands, Animal/immunology
- Mammary Glands, Animal/metabolism
- Mammary Glands, Animal/pathology
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/pathology
- Mice, Inbred BALB C
- Mice, Inbred NOD
- Mice, SCID
- Mice, Transgenic
- Natural Killer T-Cells/immunology
- Natural Killer T-Cells/metabolism
- Parity
- Pregnancy
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Signal Transduction
- Tumor Microenvironment
- Mice
Collapse
Affiliation(s)
| | - Matthew A Moss
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Mary J Feigman
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Chen Chen
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | | | | | | | - Macy Vollbrecht
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Siran Li
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Jude Kendall
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Semir Beyaz
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - John E Wilkinson
- Department of Comparative Medicine, University of Washington, Seattle, WA, USA
| | | |
Collapse
|
30
|
García-Estevez L, González-Martínez S, Moreno-Bueno G. The Leptin Axis and Its Association With the Adaptive Immune System in Breast Cancer. Front Immunol 2021; 12:784823. [PMID: 34868066 PMCID: PMC8634160 DOI: 10.3389/fimmu.2021.784823] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 10/28/2021] [Indexed: 12/21/2022] Open
Abstract
Adipose tissue secretes various peptides, including leptin. This hormone acts through the leptin receptor (Ob-R), which is expressed ubiquitously on the surface of various cells, including breast cancer cells and immune cells. Increasing evidence points to an interaction between the tumor microenvironment, tumor cells, and the immune system. Leptin plays an important role in breast cancer tumorigenesis and may be implicated in activation of the immune system. While breast cancer cannot be considered an immunogenic cancer, the triple-negative subtype is an exception. Specific immune cells - tumor infiltrating lymphocytes - are involved in the immune response and act as predictive and prognostic factors in certain breast cancer subtypes. The aim of this article is to review the interaction between adipose tissue, through the expression of leptin and its receptor, and the adaptive immune system in breast cancer.
Collapse
Affiliation(s)
- Laura García-Estevez
- Breast Cancer Department, MD Anderson Cancer Center, Madrid, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,MD Anderson International Foundation, Madrid, Spain
| | - Silvia González-Martínez
- Pathology Department, Hospital Ramón y Cajal, Madrid, Spain.,Fundación Contigo Contra el Cáncer de la Mujer, Madrid, Spain
| | - Gema Moreno-Bueno
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,MD Anderson International Foundation, Madrid, Spain.,Biochemistry Department, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas 'Alberto Sols' (CSIC-UAM), IdiPaz, Madrid, Spain
| |
Collapse
|
31
|
Kuroda H, Jamiyan T, Yamaguchi R, Kakumoto A, Abe A, Harada O, Masunaga A. Tumor microenvironment in triple-negative breast cancer: the correlation of tumor-associated macrophages and tumor-infiltrating lymphocytes. Clin Transl Oncol 2021; 23:2513-2525. [PMID: 34089486 PMCID: PMC8557183 DOI: 10.1007/s12094-021-02652-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/21/2021] [Indexed: 12/24/2022]
Abstract
PURPOSE Immune cells such as cytotoxic T cells, helper T cells, B cells or tumor-associated macrophages (TAMs) contribute to the anti-tumor response or pro-tumorigenic effect in triple negative breast cancer (TNBC). The interrelation of TAMs, T and B tumor-infiltrating lymphocytes (TILs) in TNBC has not been fully elucidated. METHODS We evaluated the association of tumor-associated macrophages, T and B TILs in TNBC. RESULTS TNBCs with a high CD68+, CD163+ TAMs and low CD4+, CD8+, CD20+ TILs had a significantly shorter relapse-free survival (RFS) and overall survival (OS) than those with low CD68+, CD163+ TAMs and high CD4+, CD8+, CD20+ TILs. TNBCs with high CD68+ TAMs/low CD8+ TILs showed a significantly shorter RFS and OS and a significantly poorer prognosis than those with high CD68+ TAMs/high CD8+ TILs, low CD68+ TAMs/high CD8+ TILs, and low CD68+/low CD8+. TNBCs with high CD163+ TAMs/low CD8+, low CD20 + TILs showed a significantly shorter RFS and OS and a significantly poorer prognosis than those with high CD163+ TAMs/high CD8+ TILs and high CD163+ TAMs /high CD20+ TILs. CONCLUSIONS Our study suggests that TAMs further create an optimal tumor microenvironment (TME) for growth and invasion of cancer cells when evasion of immunoreactions due to T and B TILs occurs. In TNBCs, all these events combine to affect prognosis. The process of TME is highly complex in TNBCs and for an improved understanding, larger validation studies are necessary to confirm these findings.
Collapse
Affiliation(s)
- H Kuroda
- Department of Diagnostic Pathology, Tokyo Women's Medical University, Medical Center East, 2-1-10 Nishiogu, Arakawa-ku, Tokyo, 116-8567, Japan.
- Department of Diagnostic Pathology, Dokkyo Medical University, Mibu, Japan.
| | - T Jamiyan
- Department of Diagnostic Pathology, Dokkyo Medical University, Mibu, Japan
- Department of Pathology and Forensic Medicine, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - R Yamaguchi
- Department of Pathology & Laboratory Medicine, Kurume University Medical Center, Kurume, Japan
| | - A Kakumoto
- Department of Diagnostic Pathology, Tokyo Women's Medical University, Medical Center East, 2-1-10 Nishiogu, Arakawa-ku, Tokyo, 116-8567, Japan
- Department of Diagnostic Pathology, Nasu Red Cross Hospital, Otawara, Japan
| | - A Abe
- Breast Center, Dokkyo Medical University, Mibu, Japan
| | - O Harada
- Breast Center, Showa University, Tokyo, Japan
| | - A Masunaga
- Department of Diagnostic Pathology, Tokyo Women's Medical University, Medical Center East, 2-1-10 Nishiogu, Arakawa-ku, Tokyo, 116-8567, Japan
| |
Collapse
|
32
|
Lu Q, Chen Z, Ji D, Mao Y, Jiang Q, Yang Z, Loor JJ. Progress on the Regulation of Ruminant Milk Fat by Noncoding RNAs and ceRNAs. Front Genet 2021; 12:733925. [PMID: 34790222 PMCID: PMC8591074 DOI: 10.3389/fgene.2021.733925] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/23/2021] [Indexed: 12/11/2022] Open
Abstract
Milk fat is not only a key factor affecting the quality of fresh milk but also a major target trait forbreeding. The regulation of milk fat involves multiple genes, network regulation and signal transduction. To explore recent discoveries of pathway regulation, we reviewed the published literature with a focus on functional noncoding RNAs and epigenetic regulation in ruminants. Results indicate that miRNAs play key roles in the regulation of milk fat synthesis and catabolism in ruminants. Although few data are available, merging evidence indicates that lncRNAs and circRNAs act on milk fat related genes through indirect action with microRNAs or RNAs in the ceRNA network to elicit positive effects on transcription. Although precise regulatory mechanisms remain unclear, most studies have focused on the regulation of the function of target genes through functional noncoding RNAs. Data to help identify factors that can regulate their own expression and function or to determine whether self-regulation involves positive and/or negative feedback are needed. Despite the growing body of research on the role of functional noncoding RNA in the control of ruminant milk fat, most data are still not translatable for field applications. Overall, the understanding of mechanisms whereby miRNA, lncRNA, circRNA, and ceRNA regulate ruminant milk fat remains an exciting area of research.
Collapse
Affiliation(s)
- QinYue Lu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education, Yangzhou University, Yangzhou, China
| | - Zhi Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education, Yangzhou University, Yangzhou, China
| | - Dejun Ji
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education, Yangzhou University, Yangzhou, China
| | - Yongjiang Mao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education, Yangzhou University, Yangzhou, China
| | - Qianming Jiang
- Mammalian Nutrition Physiology Genomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL, United States
| | - Zhangping Yang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education, Yangzhou University, Yangzhou, China
| | - Juan J Loor
- Mammalian Nutrition Physiology Genomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL, United States
| |
Collapse
|
33
|
Wilson GJ, Fukuoka A, Vidler F, Graham GJ. Diverse myeloid cells are recruited to the developing and inflamed mammary gland. Immunology 2021; 165:206-218. [PMID: 34775606 DOI: 10.1111/imm.13430] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 10/27/2021] [Accepted: 11/09/2021] [Indexed: 12/26/2022] Open
Abstract
The immune system plays fundamental roles in the mammary gland, shaping developmental processes and controlling inflammation during infection and cancer. Here, we reveal unanticipated heterogeneity in the myeloid cell compartment during development of virgin, pregnant, lactating and involuting mouse mammary glands, and in milk. We investigate the functional consequences of individual and compound chemokine receptor deficiency on cell recruitment. Diverse myeloid cell recruitment was also shown in models of sterile inflammation and bacterial infection. Strikingly, we have shown that inflammation and infection can alter the abundance of terminal end buds, a key developmental structure, within the pubertal mammary gland. This previously unknown effect of inflammatory burden during puberty could have important implications for understanding pubertal development.
Collapse
Affiliation(s)
- Gillian J Wilson
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Ayumi Fukuoka
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Francesca Vidler
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Gerard J Graham
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| |
Collapse
|
34
|
Tice RR, Bassan A, Amberg A, Anger LT, Beal MA, Bellion P, Benigni R, Birmingham J, Brigo A, Bringezu F, Ceriani L, Crooks I, Cross K, Elespuru R, Faulkner DM, Fortin MC, Fowler P, Frericks M, Gerets HHJ, Jahnke GD, Jones DR, Kruhlak NL, Lo Piparo E, Lopez-Belmonte J, Luniwal A, Luu A, Madia F, Manganelli S, Manickam B, Mestres J, Mihalchik-Burhans AL, Neilson L, Pandiri A, Pavan M, Rider CV, Rooney JP, Trejo-Martin A, Watanabe-Sailor KH, White AT, Woolley D, Myatt GJ. In Silico Approaches In Carcinogenicity Hazard Assessment: Current Status and Future Needs. COMPUTATIONAL TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2021; 20. [PMID: 35368437 DOI: 10.1016/j.comtox.2021.100191] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Historically, identifying carcinogens has relied primarily on tumor studies in rodents, which require enormous resources in both money and time. In silico models have been developed for predicting rodent carcinogens but have not yet found general regulatory acceptance, in part due to the lack of a generally accepted protocol for performing such an assessment as well as limitations in predictive performance and scope. There remains a need for additional, improved in silico carcinogenicity models, especially ones that are more human-relevant, for use in research and regulatory decision-making. As part of an international effort to develop in silico toxicological protocols, a consortium of toxicologists, computational scientists, and regulatory scientists across several industries and governmental agencies evaluated the extent to which in silico models exist for each of the recently defined 10 key characteristics (KCs) of carcinogens. This position paper summarizes the current status of in silico tools for the assessment of each KC and identifies the data gaps that need to be addressed before a comprehensive in silico carcinogenicity protocol can be developed for regulatory use.
Collapse
Affiliation(s)
- Raymond R Tice
- RTice Consulting, Hillsborough, North Carolina, 27278, USA
| | | | - Alexander Amberg
- Sanofi Preclinical Safety, Industriepark Höchst, 65926 Frankfurt, Germany
| | - Lennart T Anger
- Genentech, Inc., South San Francisco, California, 94080, USA
| | - Marc A Beal
- Healthy Environments and Consumer Safety Branch, Health Canada, Government of Canada, Ottawa, Ontario, Canada K1A 0K9
| | | | | | - Jeffrey Birmingham
- GlaxoSmithKline, David Jack Centre for R&D, Ware, Hertfordshire, SG12 0DP, United Kingdom
| | - Alessandro Brigo
- Roche Pharmaceutical Research & Early Development, Pharmaceutical Sciences, Roche Innovation, Center Basel, F. Hoffmann-La Roche Ltd, CH-4070, Basel, Switzerland
| | | | - Lidia Ceriani
- Humane Society International, 1000 Brussels, Belgium
| | - Ian Crooks
- British American Tobacco (Investments) Ltd, GR&D Centre, Southampton, SO15 8TL, United Kingdom
| | | | - Rosalie Elespuru
- Food and Drug Administration, Center for Devices and Radiological Health, Silver Spring, Maryland, 20993, USA
| | - David M Faulkner
- Chemical Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| | - Marie C Fortin
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, 08855, USA
| | - Paul Fowler
- FSTox Consulting (Genetic Toxicology), Northamptonshire, United Kingdom
| | | | | | - Gloria D Jahnke
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, 27709, USA
| | | | - Naomi L Kruhlak
- Food and Drug Administration, Center for Drug Evaluation and Research, Silver Spring, Maryland, 20993, USA
| | - Elena Lo Piparo
- Chemical Food Safety Group, Nestlé Research, CH-1000 Lausanne 26, Switzerland
| | - Juan Lopez-Belmonte
- Cuts Ice Ltd Chemical Food Safety Group, Nestlé Research, CH-1000 Lausanne 26, Switzerland
| | - Amarjit Luniwal
- North American Science Associates (NAMSA) Inc., Minneapolis, Minnesota, 55426, USA
| | - Alice Luu
- Healthy Environments and Consumer Safety Branch, Health Canada, Government of Canada, Ottawa, Ontario, Canada K1A 0K9
| | - Federica Madia
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Serena Manganelli
- Chemical Food Safety Group, Nestlé Research, CH-1000 Lausanne 26, Switzerland
| | | | - Jordi Mestres
- IMIM Institut Hospital Del Mar d'Investigacions Mèdiques and Universitat Pompeu Fabra, Doctor Aiguader 88, Parc de Recerca Biomèdica, 08003 Barcelona, Spain; and Chemotargets SL, Baldiri Reixac 4, Parc Científic de Barcelona, 08028, Barcelona, Spain
| | | | - Louise Neilson
- Broughton Nicotine Services, Oak Tree House, Earby, Lancashire, BB18 6JZ United Kingdom
| | - Arun Pandiri
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, 27709, USA
| | | | - Cynthia V Rider
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, 27709, USA
| | - John P Rooney
- Integrated Laboratory Systems, LLC., Morrisville, North Carolina, 27560, USA
| | | | - Karen H Watanabe-Sailor
- School of Mathematical and Natural Sciences, Arizona State University, West Campus, Glendale, Arizona, 85306, USA
| | - Angela T White
- GlaxoSmithKline, David Jack Centre for R&D, Ware, Hertfordshire, SG12 0DP, United Kingdom
| | | | | |
Collapse
|
35
|
Yaegashi LB, Baldavira CM, Prieto TG, Machado-Rugolo J, Velosa APP, da Silveira LKR, Assato A, Ab'Saber AM, Falzoni R, Takagaki T, Silva PL, Teodoro WR, Capelozzi VL. In Situ Overexpression of Matricellular Mechanical Proteins Demands Functional Immune Signature and Mitigates Non-Small Cell Lung Cancer Progression. Front Immunol 2021; 12:714230. [PMID: 34484217 PMCID: PMC8415570 DOI: 10.3389/fimmu.2021.714230] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/26/2021] [Indexed: 12/19/2022] Open
Abstract
Non-small cell lung carcinoma (NSCLC) is a complex cancer biome composed of malignant cells embedded in a sophisticated tumor microenvironment (TME) combined with different initiating cell types, including immune cells and cancer-associated fibroblasts (CAFs), and extracellular matrix (ECM) proteins. However, little is known about these tumors’ immune-matricellular relationship as functional and mechanical barriers. This study investigated 120 patients with NSCLC to describe the immune-matricellular phenotypes of their TME and their relationship with malignant cells. Immunohistochemistry (IHC) was performed to characterize immune checkpoints (PD-L1, LAG-3, CTLA-4+, VISTA 1), T cells (CD3+), cytotoxic T cells (CD8+, Granzyme B), macrophages (CD68+), regulatory T cells (FOXP3+, CD4+), natural killer cells (CD57+), and B lymphocytes (CD20+), whereas CAFs and collagen types I, III, and V were characterized by immunofluorescence (IF). We observed two distinct functional immune-cellular barriers—the first of which showed proximity between malignant cells and cytotoxic T cells, and the second of which showed distant proximity between non-cohesive nests of malignant cells and regulatory T cells. We also identified three tumor-associated matricellular barriers: the first, with a localized increase in CAFs and a low deposition of Col V, the second with increased CAFs, Col III and Col I fibers, and the third with a high amount of Col fibers and CAFs bundled and aligned perpendicularly to the tumor border. The Cox regression analysis was designed in two steps. First, we investigated the relationship between the immune-matricellular components and tumor pathological stage (I, II, and IIIA), and better survival rates were seen in patients whose tumors expressed collagen type III > 24.89 fibers/mm². Then, we included patients who had progressed to pathological stage IV and found an association between poor survival and tumor VISTA 1 expression > 52.86 cells/mm² and CD3+ ≤ 278.5 cells/mm². We thus concluded that differential patterns in the distribution of immune-matricellular phenotypes in the TME of NSCLC patients could be used in translational studies to predict new treatment strategies and improve patient outcome. These data raise the possibility that proteins with mechanical barrier function in NSCLC may be used by cancer cells to protect them from immune cell infiltration and immune-mediated destruction, which can otherwise be targeted effectively with immunotherapy or collagen therapy.
Collapse
Affiliation(s)
| | | | | | - Juliana Machado-Rugolo
- Department of Pathology, University of São Paulo Medical School, São Paulo, Brazil.,Health Technology Assessment Center (NATS), Clinical Hospital (HCFMB), Medical School of São Paulo State University (UNESP), Botucatu, Brazil
| | - Ana Paula Pereira Velosa
- Rheumatology Division of the Hospital das Clinicas, University of São Paulo Medical School, São Paulo, Brazil
| | | | - Aline Assato
- Department of Pathology, University of São Paulo Medical School, São Paulo, Brazil
| | | | - Roberto Falzoni
- Department of Pathology, University of São Paulo Medical School, São Paulo, Brazil
| | - Teresa Takagaki
- Division of Pneumology, Instituto do Coração (Incor), University of São Paulo Medical School (USP), São Paulo, Brazil
| | - Pedro Leme Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Rio de Janeiro, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| | - Walcy Rosolia Teodoro
- Rheumatology Division of the Hospital das Clinicas, University of São Paulo Medical School, São Paulo, Brazil
| | - Vera Luiza Capelozzi
- Department of Pathology, University of São Paulo Medical School, São Paulo, Brazil
| |
Collapse
|
36
|
Stokowa-Sołtys K, Wojtkowiak K, Jagiełło K. Fusobacterium nucleatum - Friend or foe? J Inorg Biochem 2021; 224:111586. [PMID: 34425476 DOI: 10.1016/j.jinorgbio.2021.111586] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 08/13/2021] [Accepted: 08/15/2021] [Indexed: 01/16/2023]
Abstract
Fusobacterium nucleatum (F. nucleatum) is one of the most abundant Gram-negative anaerobic bacteria, part of the gut, and oral commensal flora, generally found in human dental plaque. Its presence could be associated with various human diseases, including, e.g., periodontal, angina, lung and gynecological abscesses. This bacteria can enter the blood circulation as a result of periodontal infection. It was proven that F. nucleatum migrates from its primary site of colonization in the oral cavity to other parts of the body. It could cause numerous diseases, including cancers. On the other hand, it was shown that Fusobacterium produces significant amounts of butyric acid, which is a great source of energy for colonocytes (anti-inflammatory cells). Therefore, it is very interesting to get to know the two faces of F. nucleatum.
Collapse
Affiliation(s)
- Kamila Stokowa-Sołtys
- Faculty of Chemistry, University of Wroclaw, F. Joliot-Curie 14, 50-383 Wroclaw, Poland.
| | - Kamil Wojtkowiak
- Faculty of Chemistry, University of Wroclaw, F. Joliot-Curie 14, 50-383 Wroclaw, Poland
| | - Karolina Jagiełło
- Faculty of Chemistry, University of Wroclaw, F. Joliot-Curie 14, 50-383 Wroclaw, Poland
| |
Collapse
|
37
|
Wang DK, Zuo Q, He QY, Li B. Targeted Immunotherapies in Gastrointestinal Cancer: From Molecular Mechanisms to Implications. Front Immunol 2021; 12:705999. [PMID: 34447376 PMCID: PMC8383067 DOI: 10.3389/fimmu.2021.705999] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 07/26/2021] [Indexed: 12/12/2022] Open
Abstract
Gastrointestinal cancer is a leading cause of cancer-related mortality and remains a major challenge for cancer treatment. Despite the combined administration of modern surgical techniques and chemoradiotherapy (CRT), the overall 5-year survival rate of gastrointestinal cancer patients in advanced stage disease is less than 15%, due to rapid disease progression, metastasis, and CRT resistance. A better understanding of the mechanisms underlying cancer progression and optimized treatment strategies for gastrointestinal cancer are urgently needed. With increasing evidence highlighting the protective role of immune responses in cancer initiation and progression, immunotherapy has become a hot research topic in the integrative management of gastrointestinal cancer. Here, an overview of the molecular understanding of colorectal cancer, esophageal cancer and gastric cancer is provided. Subsequently, recently developed immunotherapy strategies, including immune checkpoint inhibitors, chimeric antigen receptor T cell therapies, tumor vaccines and therapies targeting other immune cells, have been described. Finally, the underlying mechanisms, fundamental research and clinical trials of each agent are discussed. Overall, this review summarizes recent advances and future directions for immunotherapy for patients with gastrointestinal malignancies.
Collapse
Affiliation(s)
| | | | | | - Bin Li
- Ministry of Education (MOE), Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, Jinan University, Guangzhou, China
| |
Collapse
|
38
|
Singh K, Yadav D, Jain M, Singh PK, Jin JO. Immunotherapy for the Breast Cancer treatment: Current Evidence and Therapeutic Options. Endocr Metab Immune Disord Drug Targets 2021; 22:212-224. [PMID: 33902424 DOI: 10.2174/1871530321666210426125904] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 03/04/2021] [Accepted: 03/08/2021] [Indexed: 11/22/2022]
Abstract
Breast cancer (BC) stands at the first position among all forms of malignancies found in women globally. The available therapeutic approaches for breast cancer includes chemotherapy, radiation therapy, hormonal therapy and finally surgery. Despite the conventional therapies, in recent years the advance immunology based therapeutics emerge a potential in breast cancer treatment, including immune checkpoint blockades, vaccines and in combination with other treatment strategies. Although, commonly used treatments like trastuzumab/pertuzumab for human epidermal growth factor receptor 2 (Her2) positive and hormone therapy for estrogen receptor (ER) positive and/or progesterone receptor (PR) positive BC are specific but triple negative breast cancer (TNBC) cases remain a great challenge for treatment measures. Immune checkpoint inhibitors (anti-PD-1/ anti-CTLA-4) and anti-cancer vaccines (NeuVax, Muc-1, AVX901, INO-1400 and CEA), either alone or in combination with other therapies have created new paradigm in therapeutic world. In this review, we highlighted the current immunotherapeutic aspects and their ongoing trials towards the better treatment regimen for BC.
Collapse
Affiliation(s)
- Kavita Singh
- Centre for Translational Research, School of Studies in Biochemistry, Jiwaji University, Gwalior-474011, Madhya Pradesh, India
| | - Dhananjay Yadav
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 712-749, South Korea
| | - Meenu Jain
- ICMR-AMR Diagnostics Taskforce, ECD Division, Indian Council of Medical research, Ansari Nagar, New Delhi-110029, India
| | - Pramod Kumar Singh
- Department of Biosciences, Christian Eminent College, Indore, (MP), India
| | - Jun-O Jin
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai 201508, China
| |
Collapse
|
39
|
Wang X, Jia Y, Wen L, Mu W, Wu X, Liu T, Liu X, Fang J, Luan Y, Chen P, Gao J, Nguyen KA, Cui J, Zeng G, Lan P, Chen Q, Cheng B, Wang Z. Porphyromonas gingivalis Promotes Colorectal Carcinoma by Activating the Hematopoietic NLRP3 Inflammasome. Cancer Res 2021; 81:2745-2759. [PMID: 34003774 DOI: 10.1158/0008-5472.can-20-3827] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/11/2021] [Accepted: 03/16/2021] [Indexed: 11/16/2022]
Abstract
Porphyromonas gingivalis (P. gingivalis) is a keystone periodontal pathogen associated with various digestive cancers. However, whether P. gingivalis can promote colorectal cancer and the underlying mechanism associated with such promotion remains unclear. In this study, we found that P. gingivalis was enriched in human feces and tissue samples from patients with colorectal cancer compared with those from patients with colorectal adenoma or healthy subjects. Cohort studies demonstrated that P. gingivalis infection was associated with poor prognosis in colorectal cancer. P. gingivalis increased tumor counts and tumor volume in the ApcMin/+ mouse model and increased tumor growth in orthotopic rectal and subcutaneous carcinoma models. Furthermore, orthotopic tumors from mice exposed to P. gingivalis exhibited tumor-infiltrating myeloid cell recruitment and a proinflammatory signature. P. gingivalis promoted colorectal cancer via NLRP3 inflammasome activation in vitro and in vivo. NLRP3 chimeric mice harboring orthotopic tumors showed that the effect of NLRP3 on P. gingivalis pathogenesis was mediated by hematopoietic sources. Collectively, these data suggest that P. gingivalis contributes to colorectal cancer neoplasia progression by activating the hematopoietic NLRP3 inflammasome. SIGNIFICANCE: This study demonstrates that the periodontal pathogen P. gingivalis can promote colorectal tumorigenesis by recruiting myeloid cells and creating a proinflammatory tumor microenvironment. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/10/2745/F1.large.jpg.
Collapse
Affiliation(s)
- Xi Wang
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Yiqun Jia
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China.,Stomatology Center, Shenzhen People's Hospital, the Second Clinical Medical College of Jinan University, the First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Liling Wen
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Wenxin Mu
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Xianrui Wu
- Department of Colorectal Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tao Liu
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xiangqi Liu
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Juan Fang
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Yizhao Luan
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Lab of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Ping Chen
- Department of Gastroenterology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Jinlong Gao
- Institute of Dental Research, Sydney Dental School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Ky-Anh Nguyen
- Institute of Dental Research, Sydney Dental School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Jun Cui
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Gucheng Zeng
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Ping Lan
- Department of Colorectal Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qianming Chen
- The Affiliated Hospital of Stomatology, and Key Laboratory of Oral Biomedical Research of Zhejiang Province, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Bin Cheng
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China.
| | - Zhi Wang
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
40
|
Targeting the purinergic pathway in breast cancer and its therapeutic applications. Purinergic Signal 2021; 17:179-200. [PMID: 33576905 PMCID: PMC7879595 DOI: 10.1007/s11302-020-09760-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 12/06/2020] [Indexed: 12/21/2022] Open
Abstract
Breast cancer (BC) is the most frequent cause of death among women, representing a global public health problem. Here, we aimed to discuss the correlation between the purinergic system and BC, recognizing therapeutic targets. For this, we analyzed the interaction of extracellular nucleotides and nucleosides with the purinergic receptors P1 and P2, as well as the influence of ectonucleotidase enzymes (CD39 and CD73) on tumor progression. A comprehensive bibliographic search was carried out. The relevant articles for this review were found in the PubMed, Scielo, Lilacs, and ScienceDirect databases. It was observed that among the P1 receptors, the A1, A2A, and A2B receptors are involved in the proliferation and invasion of BC, while the A3 receptor is related to the inhibition of tumor growth. Among the P2 receptors, the P2X7 has a dual function. When activated for a short time, it promotes metastasis, but when activated for long periods, it is related to BC cell death. P2Y2 and P2Y6 receptors are related to BC proliferation and invasiveness. Also, the high expression of CD39 and CD73 in BC is strongly related to a worse prognosis. The receptors and ectonucleotidases involved with BC become possible therapeutic targets. Several purinergic pathways have been found to be involved in BC cell survival and progression. In this review, in addition to analyzing the pathways involved, we reviewed the therapeutic interventions already studied for BC related to the purinergic system, as well as to other possible therapeutic targets.
Collapse
|
41
|
HDAC inhibition potentiates anti-tumor activity of macrophages and enhances anti-PD-L1-mediated tumor suppression. Oncogene 2021; 40:1836-1850. [PMID: 33564072 PMCID: PMC7946638 DOI: 10.1038/s41388-020-01636-x] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 12/11/2020] [Accepted: 12/17/2020] [Indexed: 01/30/2023]
Abstract
Despite the widespread use of the blockade of immune checkpoints, for a significant number of cancer patients, these therapies have proven ineffective, presumably due to the immunosuppressive nature of the tumor microenvironment (TME). Critical drivers of immune escape in the TME include tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs), which not only mediate immune suppression, but also facilitate metastatic dissemination and impart resistance to immunotherapies. Thus, strategies that convert them into tumor fighters may offer great therapeutic potential. In this study, we evaluated whether pharmacologic modulation of macrophage phenotype by HDAC inhibitors (HDACi) could produce an anti-tumor effect. We demonstrated that low-dose HDACi trichostatin-A (TSA) markedly reshaped the tumor immune microenvironment by modulating the suppressive activity of infiltrating macrophages and inhibiting the recruitment of MDSCs in various tumors. These actions, in turn, augmented anti-tumor immune responses and further enhanced anti-tumor effects of immunotherapies. HDAC inhibition, however, also upregulated PD-L1, thereby limiting the beneficial therapeutic effects. Indeed, combining low-dose TSA with anti-PD-L1 in this model significantly enhanced the durability of tumor reduction and prolonged survival of tumor-bearing mice, compared with the effect of either treatment alone. These data introduce HDAC inhibition as a potential means to harness the anti-tumor potential of macrophages in cancer therapy.
Collapse
|
42
|
Prognostic value of tumor-infiltrating lymphocyte subtypes in residual tumors of patients with triple-negative breast cancer after neoadjuvant chemotherapy. Chin Med J (Engl) 2020; 133:552-560. [PMID: 32044815 PMCID: PMC7065871 DOI: 10.1097/cm9.0000000000000656] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background: After neoadjuvant chemotherapy (NAC), non-pathological complete response of breast cancer patients can benefit from tailored adjuvant chemotherapy. However, it is difficult to select patients with poorer prognosis for additional adjuvant chemotherapy to maximize the benefits. Our study aimed to explore whether the subtypes of tumor-infiltrating lymphocytes (TILs) in residual tumors (RT) is related to the prognosis of triple-negative breast cancer (TNBC) after NAC. Methods: Data from patients with primary TNBC consecutively diagnosed at the Breast Disease Center of Peking University First Hospital from 2008 to 2014 were retrieved, and the cases with RT in the breast after NAC were enrolled. TILs subtypes in RT were observed by double-staining immunohistochemistry, and counted with the median TILs value per square millimeter as the cut-off to define high versus low TILs density in each subtype. The relationships between the TIL density of each subgroup and the clinicopathological characteristics of the RT after NAC patients were analyzed by Fisher exact test. Disease-free survival (DFS) and overall survival (OS) were analyzed by the Kaplan-Meier method and log-rank statistics. Results: A total of 37 eligible patients were included in this study, and the median follow-up period was 50 months (range 17–106 months). There was no significant correlation between the infiltrate density of CD4+, CD8+, CD20+, and CD68+ lymphocytes and clinic-pathological characteristics. Significantly better prognosis was observed in patients with high CD4+-TILs (DFS: P = 0.005, OS: P = 0.021) and high CD8+-TILs (DFS: P = 0.018) and low CD20+-TILs (OS: P = 0.042). Further analysis showed that patients with CD4+/CD20+ ratio greater than 1 (DFS: P = 0.001, OS: P = 0.002) or CD8+/CD20+ ratio greater than 1 (DFS: P = 0.009, OS: P = 0.022) had a better prognosis. Conclusions: Subtypes of TILs in RT is a potential predictive biomarker of survival in TNBC patients after NAC.
Collapse
|
43
|
Watson CJ, Khaled WT. Mammary development in the embryo and adult: new insights into the journey of morphogenesis and commitment. Development 2020; 147:dev169862. [PMID: 33191272 DOI: 10.1242/dev.169862] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The mammary gland is a unique tissue and the defining feature of the class Mammalia. It is a late-evolving epidermal appendage that has the primary function of providing nutrition for the young, although recent studies have highlighted additional benefits of milk including the provision of passive immunity and a microbiome and, in humans, the psychosocial benefits of breastfeeding. In this Review, we outline the various stages of mammary gland development in the mouse, with a particular focus on lineage specification and the new insights that have been gained by the application of recent technological advances in imaging in both real-time and three-dimensions, and in single cell RNA sequencing. These studies have revealed the complexity of subpopulations of cells that contribute to the mammary stem and progenitor cell hierarchy and we suggest a new terminology to distinguish these cells.
Collapse
Affiliation(s)
- Christine J Watson
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| | - Walid T Khaled
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| |
Collapse
|
44
|
Wilsher MJ, Banerjee D. Eosinophilic mastitis in a lactating breast. Pathology 2020; 53:549-550. [PMID: 33168146 DOI: 10.1016/j.pathol.2020.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/27/2020] [Accepted: 08/03/2020] [Indexed: 10/23/2022]
Affiliation(s)
| | - Dibyesh Banerjee
- Breast Unit, Rose Centre, St George's University Hospital, London, UK
| |
Collapse
|
45
|
Prolactin: A hormone with diverse functions from mammary gland development to cancer metastasis. Semin Cell Dev Biol 2020; 114:159-170. [PMID: 33109441 DOI: 10.1016/j.semcdb.2020.10.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/04/2020] [Accepted: 10/11/2020] [Indexed: 01/14/2023]
Abstract
Prolactin has a rich mechanistic set of actions and signaling in order to elicit developmental effects in mammals. Historically, prolactin has been appreciated as an endocrine peptide hormone that is responsible for final, functional mammary gland development and lactation. Multiple signaling pathways impacted upon by the microenvironment contribute to cell function and differentiation. Endocrine, autocrine and paracrine signaling are now apparent in not only mammary development, but also in cancer, and involve multiple cell types including those of the immune system. Multiple ligands agonists are capable of binding to the prolactin receptor, potentially expanding receptor function. Prolactin has an important role not only in tumorigenesis of the breast, but also in a number of hormonally responsive cancers such as prostate, ovarian and endometrial cancer, as well as pancreatic and lung cancer. Although pituitary and extra-pituitary sources of prolactin such as the epithelium are important, stromal sourced prolactin is now also being recognized as an important factor in tumor progression, all of which potentially signal to multiple cell types in the tumor microenvironment. While prolactin has important roles in milk production including calcium and bone homeostasis, in the disease state it can also affect bone homeostasis. Prolactin also impacts metastatic cancer of the breast to modulate the bone microenvironment and promote bone damage. Prolactin has a fascinating contribution in both physiologic and pathologic settings of mammals.
Collapse
|
46
|
Prevalence and clinical relevance of tumor-associated tissue eosinophilia (TATE) in breast cancer. Surgery 2020; 169:1234-1239. [PMID: 32958266 DOI: 10.1016/j.surg.2020.07.052] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 07/20/2020] [Accepted: 01/12/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Tumor-associated tissue eosinophilia (TATE) has been associated with outcomes in a variety of solid tumors; however, its role in breast cancer is not well defined. We hypothesized that tumor-associated tissue eosinophilia is associated with a high mutation and neoantigen load, and we assessed its correlation with cancer outcomes. METHODS The Cancer Genome Atlas was analyzed for eosinophil signatures in breast cancer specimens. Descriptive analyses were performed, including the tumor-infiltrating cell composition using CIBERSORT, cytolytic activity score, and gene set enrichment analysis. Overall survival and disease-free survival were calculated using the Kaplan-Meier method. RESULTS Out of 1069 cases analyzed, 40 (3.7%) had tissue eosinophils (the tumor-associated tissue eosinophilia group). Tumor-associated tissue eosinophilia was noted in 32.5% luminal, 5% HER2-positive, and 15% triple-negative breast cancer subtypes. The single nucleotide variant-neoantigen load was significantly higher in the tumor-associated tissue eosinophilia group (P = .005), with a higher nonsilent mutation rate (P = .01). The tumor-associated tissue eosinophilia group had lower cytolytic activity (P = .02) but had enriched MYC-targeted (P = .002), E2F-targeted (P = .04), deoxyribonucleic acid repair (P = .03), and unfolded protein response gene sets (P = .05). Tumor-associated tissue eosinophilia was associated with a trend toward improved disease-free survival (P = .06) but presented no differences in overall survival (P = .56). CONCLUSION Tumor-associated tissue eosinophilia was noted in 3.7% of breast cancers and was associated with a higher single nucleotide variant-neoantigen load and nonsilent mutation rate, similar to that of tumor-infiltrating lymphocytes in the triple-negative subtype. However, a lower cytolytic activity score and enriched cell proliferation-related gene sets implicate different roles for tumor-associated tissue eosinophilia than for tumor-infiltrating lymphocytes.
Collapse
|
47
|
Myeloid Cell Modulation by Tumor-Derived Extracellular Vesicles. Int J Mol Sci 2020; 21:ijms21176319. [PMID: 32878277 PMCID: PMC7504548 DOI: 10.3390/ijms21176319] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/28/2020] [Accepted: 08/29/2020] [Indexed: 12/17/2022] Open
Abstract
Extracellular vesicles (EV) can carry proteins, RNA and DNA, thus serving as communication tools between cells. Tumor cells secrete EV, which can be taken up by surrounding cells in the tumor microenvironment as well as by cells in distant organs. Tumor-derived EV (TEV) contain factors induced by tumor-associated hypoxia such as heat shock proteins or a variety of microRNA (miRNA). The interaction of TEV with tumor and host cells can promote cancer angiogenesis, invasion and metastasis. Myeloid cells are widely presented in tissues, comprise the majority of immune cells and play an essential role in immune reactions and tissue remodeling. However, in cancer, the differentiation of myeloid cells and their functions are impaired, resulting in tumor promotion. Such alterations are due to chronic inflammatory conditions associated with cancer and are mediated by the tumor secretome, including TEV. A high capacity of myeloid cells to clear EV from circulation put them in the central position in EV-mediated formation of pre-metastatic niches. The exposure of myeloid cells to TEV could trigger numerous signaling pathways. Progenitors of myeloid cells alter their differentiation upon the contact with TEV, resulting in the generation of myeloid-derived suppressor cells (MDSC), inhibiting anti-tumor function of T and natural killer (NK) cells and promoting thereby tumor progression. Furthermore, TEV can augment MDSC immunosuppressive capacity. Different subsets of mature myeloid cells such as monocytes, macrophages, dendritic cells (DC) and granulocytes take up TEV and acquire a protumorigenic phenotype. However, the delivery of tumor antigens to DC by TEV was shown to enhance their immunostimulatory capacity. The present review will discuss a diverse and complex EV-mediated crosstalk between tumor and myeloid cells in the context of the tumor type, TEV-associated cargo molecules and type of recipient cells.
Collapse
|
48
|
Wilson GJ, Fukuoka A, Love SR, Kim J, Pingen M, Hayes AJ, Graham GJ. Chemokine receptors coordinately regulate macrophage dynamics and mammary gland development. Development 2020; 147:dev187815. [PMID: 32467242 PMCID: PMC7328164 DOI: 10.1242/dev.187815] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 05/20/2020] [Indexed: 12/22/2022]
Abstract
Macrophages are key regulators of developmental processes, including those involved in mammary gland development. We have previously demonstrated that the atypical chemokine receptor ACKR2 contributes to the control of ductal epithelial branching in the developing mammary gland by regulating macrophage dynamics. ACKR2 is a chemokine-scavenging receptor that mediates its effects through collaboration with inflammatory chemokine receptors (iCCRs). Here, we reveal reciprocal regulation of branching morphogenesis in the mammary gland, whereby stromal ACKR2 modulates levels of the shared ligand CCL7 to control the movement of a key population of CCR1-expressing macrophages to the ductal epithelium. In addition, oestrogen, which is essential for ductal elongation during puberty, upregulates CCR1 expression on macrophages. The age at which girls develop breasts is decreasing, which raises the risk of diseases including breast cancer. This study presents a previously unknown mechanism controlling the rate of mammary gland development during puberty and highlights potential therapeutic targets.
Collapse
MESH Headings
- Animals
- Chemokine CCL3/deficiency
- Chemokine CCL3/genetics
- Chemokine CCL3/metabolism
- Chemokine CCL5/deficiency
- Chemokine CCL5/genetics
- Chemokine CCL5/metabolism
- Epithelium/metabolism
- Estradiol/pharmacology
- Female
- Lectins, C-Type/metabolism
- Macrophages/cytology
- Macrophages/metabolism
- Mammary Glands, Animal/growth & development
- Mammary Glands, Animal/metabolism
- Mannose Receptor
- Mannose-Binding Lectins/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Morphogenesis
- Receptors, CCR1/deficiency
- Receptors, CCR1/genetics
- Receptors, CCR1/metabolism
- Receptors, Cell Surface/metabolism
- Receptors, Chemokine/deficiency
- Receptors, Chemokine/genetics
- Receptors, Chemokine/metabolism
- Up-Regulation/drug effects
Collapse
Affiliation(s)
- Gillian J Wilson
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK
| | - Ayumi Fukuoka
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK
| | - Samantha R Love
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK
| | - Jiwon Kim
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK
- Department of Physiology, University of Toronto, Medical Sciences Building, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Marieke Pingen
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK
| | - Alan J Hayes
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK
| | - Gerard J Graham
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK
| |
Collapse
|
49
|
Domingo-Gonzalez R, Zanini F, Che X, Liu M, Jones RC, Swift MA, Quake SR, Cornfield DN, Alvira CM. Diverse homeostatic and immunomodulatory roles of immune cells in the developing mouse lung at single cell resolution. eLife 2020; 9:e56890. [PMID: 32484158 PMCID: PMC7358008 DOI: 10.7554/elife.56890] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 05/13/2020] [Indexed: 12/20/2022] Open
Abstract
At birth, the lungs rapidly transition from a pathogen-free, hypoxic environment to a pathogen-rich, rhythmically distended air-liquid interface. Although many studies have focused on the adult lung, the perinatal lung remains unexplored. Here, we present an atlas of the murine lung immune compartment during early postnatal development. We show that the late embryonic lung is dominated by specialized proliferative macrophages with a surprising physical interaction with the developing vasculature. These macrophages disappear after birth and are replaced by a dynamic mixture of macrophage subtypes, dendritic cells, granulocytes, and lymphocytes. Detailed characterization of macrophage diversity revealed an orchestration of distinct subpopulations across postnatal development to fill context-specific functions in tissue remodeling, angiogenesis, and immunity. These data both broaden the putative roles for immune cells in the developing lung and provide a framework for understanding how external insults alter immune cell phenotype during a period of rapid lung growth and heightened vulnerability.
Collapse
Affiliation(s)
- Racquel Domingo-Gonzalez
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
- Center for Excellence in Pulmonary Biology, Stanford University School of MedicineStanfordUnited States
| | - Fabio Zanini
- Department of Bioengineering, Stanford UniversityStanfordUnited States
- Prince of Wales Clinical School, Lowy Cancer Research Centre, University of New South WalesSydneyAustralia
| | - Xibing Che
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
- Center for Excellence in Pulmonary Biology, Stanford University School of MedicineStanfordUnited States
- Division of Pulmonary, Asthma and Sleep Medicine, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
| | - Min Liu
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
- Center for Excellence in Pulmonary Biology, Stanford University School of MedicineStanfordUnited States
| | - Robert C Jones
- Department of Bioengineering, Stanford UniversityStanfordUnited States
| | - Michael A Swift
- Department of Chemical and Systems Biology, Stanford UniversityStanfordUnited States
| | - Stephen R Quake
- Department of Bioengineering, Stanford UniversityStanfordUnited States
- Chan Zuckerberg BiohubSan FranciscoUnited States
- Department of Applied Physics, Stanford UniversityStanfordUnited States
| | - David N Cornfield
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
- Center for Excellence in Pulmonary Biology, Stanford University School of MedicineStanfordUnited States
- Division of Pulmonary, Asthma and Sleep Medicine, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
| | - Cristina M Alvira
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
- Center for Excellence in Pulmonary Biology, Stanford University School of MedicineStanfordUnited States
| |
Collapse
|
50
|
Microbial Alterations and Risk Factors of Breast Cancer: Connections and Mechanistic Insights. Cells 2020; 9:cells9051091. [PMID: 32354130 PMCID: PMC7290701 DOI: 10.3390/cells9051091] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/21/2020] [Accepted: 04/23/2020] [Indexed: 12/12/2022] Open
Abstract
Breast cancer-related mortality remains high worldwide, despite tremendous advances in diagnostics and therapeutics; hence, the quest for better strategies for disease management, as well as the identification of modifiable risk factors, continues. With recent leaps in genomic technologies, microbiota have emerged as major players in most cancers, including breast cancer. Interestingly, microbial alterations have been observed with some of the established risk factors of breast cancer, such as obesity, aging and periodontal disease. Higher levels of estrogen, a risk factor for breast cancer that cross-talks with other risk factors such as alcohol intake, obesity, parity, breastfeeding, early menarche and late menopause, are also modulated by microbial dysbiosis. In this review, we discuss the association between known breast cancer risk factors and altered microbiota. An important question related to microbial dysbiosis and cancer is the underlying mechanisms by which alterations in microbiota can support cancer progression. To this end, we review the involvement of microbial metabolites as effector molecules, the modulation of the metabolism of xenobiotics, the induction of systemic immune modulation, and altered responses to therapy owing to microbial dysbiosis. Given the association of breast cancer risk factors with microbial dysbiosis and the multitude of mechanisms altered by dysbiotic microbiota, an impaired microbiome is, in itself, an important risk factor.
Collapse
|