1
|
Tilg H, Ianiro G, Gasbarrini A, Adolph TE. Adipokines: masterminds of metabolic inflammation. Nat Rev Immunol 2025; 25:250-265. [PMID: 39511425 DOI: 10.1038/s41577-024-01103-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2024] [Indexed: 11/15/2024]
Abstract
Adipose tissue is an immunologically active organ that controls host physiology, partly through the release of mediators termed adipokines. In obesity, adipocytes and infiltrating leukocytes produce adipokines, which include the hormones adiponectin and leptin and cytokines such as tumour necrosis factor and IL-1β. These adipokines orchestrate immune responses that are collectively referred to as metabolic inflammation. Consequently, metabolic inflammation characterizes metabolic disorders and promotes distinct disease aspects, such as insulin resistance, metabolic dysfunction-associated liver disease and cardiovascular complications. In this unifying concept, adipokines participate in the immunological cross-talk that occurs between metabolically active organs in metabolic diseases, highlighting the fundamental role of adipokines in obesity and their potential for therapeutic intervention. Here, we summarize how adipokines shape metabolic inflammation in mice and humans, focusing on their contribution to metabolic disorders in the setting of obesity and discussing their value as therapeutic targets.
Collapse
Affiliation(s)
- Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| | - Gianluca Ianiro
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Antonio Gasbarrini
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
2
|
Neamah AS, Wadan AHS, Lafta FM, Elakwa DES. The potential role of targeting the leptin receptor as a treatment for breast cancer in the context of hyperleptinemia: a literature review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:3451-3466. [PMID: 39565396 DOI: 10.1007/s00210-024-03592-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 10/31/2024] [Indexed: 11/21/2024]
Abstract
Since cancer is becoming a leading cause of death worldwide, efforts should be concentrated on understanding its underlying biological alterations that would be utilized in disease management, especially prevention strategies. Within this context, multiple bodies of evidence have highlighted leptin's practical and promising role, a peptide hormone extracted from adipose and fatty tissues with other adipokines, in promoting the proliferation, migration, and metastatic invasion of breast carcinoma cells. Excessive blood leptin levels and hyperleptinemia increase body fat content and stimulate appetite. Also, high leptin level is believed to be associated with several conditions, including overeating, emotional stress, inflammation, obesity, and gestational diabetes. It has been noted that when leptin has impaired signaling in CNS, causing the lack of its normal function in energy balance, it results in leptin resistance, leading to a rise in its concentration in peripheral tissues. Our research paper will shed highlighting on potentially targeting the leptin receptor and its cellular signaling in suppressing breast cancer progression.
Collapse
Affiliation(s)
- Abbas S Neamah
- Department of Biology, College of Sciences, University of Baghdad, Baghdad, Iraq.
| | - Al-Hassan Soliman Wadan
- Oral Biology Department, Faculty of Dentistry, Galala University, Galala Plateau, Attaka, Suez Governorate, 15888, Egypt
| | - Fadhel M Lafta
- Department of Biology, College of Sciences, University of Baghdad, Baghdad, Iraq
| | - Doha El-Sayed Elakwa
- Department of Biochemistry & Molecular Biology, Faculty of Pharmacy for Girls, Al-Azhar University, Cairo, Egypt
- Department of Biochemistry, Faculty of Pharmacy, Sinai University, Kantra Branch, Ismailia, Egypt
| |
Collapse
|
3
|
Sagliocchi S, Acampora L, Barone B, Crocetto F, Dentice M. The impact of the tumor microenvironment in the dual burden of obesity-cancer link. Semin Cancer Biol 2025; 112:36-42. [PMID: 40127706 DOI: 10.1016/j.semcancer.2025.03.001] [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: 01/30/2025] [Revised: 03/06/2025] [Accepted: 03/11/2025] [Indexed: 03/26/2025]
Abstract
Obesity induces systemic perturbations of tissue homeostasis, leading to dyslipidemia, insulin resistance and chronic state of inflammation. Evidence from clinical and preclinical studies links excess of adiposity with increased cancer incidence and suggests that chronic inflammation may contribute to increased cancer risk in obese patients. Over the last decades of obesity research, multifaced and complicated effects of abnormal or excessive expansion of Adipose Tissue have been uncovered. In particular, it is widely described how obesity can exacerbate the tumorigenesis for instance by fueling soluble signals and adipokines and by enhancing tissue inflammation and altering the hormonal balance. Less is known about the paracrine effects of the cancer-associated adipocytes on the tumor cells and still poorly explored is the reciprocal communication between cancer cells and the adipose component of the tumor microenvironment (TME). In this review, we will address the mechanisms by which the peritumoral Adipose Tissue can influence the dynamics of tumoral cells. We will discuss how obesity-induced changes in the tumor microenvironment may enhance tumor growth and aggressive characteristics leading to increased invasiveness and metastatic progression of cancer that leads to a worsen cancer survival in obese subjects. We conclude that targeting the peritumoral adipose component of the TME would be a therapeutic option to prevent cancer development.
Collapse
Affiliation(s)
- Serena Sagliocchi
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Lucia Acampora
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Biagio Barone
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", Naples 80131, Italy
| | - Felice Crocetto
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", Naples 80131, Italy
| | - Monica Dentice
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy; CEINGE - Biotecnologie Avanzate Scarl, Naples, Italy.
| |
Collapse
|
4
|
Mane N, Fouqani A, Mrah S, Omari M, Bouaddi O, Faure E, El Fahime EM, Lkhoyaali S, Boutayeb S, El Rhazi K, Nejjari C, Huybrechts I, Khalis M. Obesity and Risk of Pre- and Postmenopausal Breast Cancer in Africa: A Systematic Review. Curr Oncol 2025; 32:167. [PMID: 40136371 PMCID: PMC11941656 DOI: 10.3390/curroncol32030167] [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: 01/26/2025] [Revised: 03/10/2025] [Accepted: 03/13/2025] [Indexed: 03/27/2025] Open
Abstract
Background and Aims: Several epidemiological studies have investigated the relationship between anthropometric factors and breast cancer (BC), but the results, particularly for premenopausal BC, remain inconsistent and contradictory. The aim of this systematic review is to present an overview of studies examining the association between obesity and BC risk in African women, by menopausal status. Methods: PubMed, Scopus, Web of Science, and Google Scholar were searched until 17 February 2025 to identify published articles. The review included original studies, with no restrictions on publication date or language. The exposures studied were height, weight, body mass index (BMI), waist circumference (WC), hip circumference (HC), and waist-to-hip ratio (WHR). The quality of the studies was assessed using the National Institute of Health (NIH). Study selection and data extraction were carried out by two authors separately. Results: A total of fifteen case-control studies were included in this systematic review, comprising 45,056 subjects (7221 cases and 37,835 controls). Among them, fourteen studies reported stratified results for pre- and postmenopausal women, and one reported findings for only premenopausal BC. We found that BMI was associated with an increased risk of BC in both premenopausal and postmenopausal women, though the associations varied across studies. Height was associated with an increased risk of pre- and postmenopausal BC. WHR was positively associated with BC in pre- and postmenopausal women, while WC showed a positive association with the risk of postmenopausal BC, and inconsistent results with premenopausal BC. Finally, a higher HC was positively associated with premenopausal and postmenopausal BC. Conclusions: The risk of developing BC is higher in obese postmenopausal women. The protective role of BMI has not been demonstrated in African premenopausal women. WHR is a risk factor for premenopausal and postmenopausal BC. There is a need to study the influence of stages of overweight and obesity on BC risk in a large sample of African women in-depth.
Collapse
Affiliation(s)
- Najia Mane
- Laboratory of Epidemiology and Research in Health Sciences, Faculty of Medicine, Pharmacy and Dental Medicine, Sidi Mohamed Ben Abdallah University, Fez 30070, Morocco; (M.O.); (K.E.R.); (C.N.)
- Department of Public Health and Clinical Research, Mohammed VI Center for Research and Innovation, Rabat 10112, Morocco; (O.B.); (E.M.E.F.); (S.L.); (S.B.)
| | - Aya Fouqani
- Faculty of Medicine and Pharmacy of Rabat, Mohamed V University in Rabat, Rabat 10000, Morocco;
| | - Siham Mrah
- Laboratory Research of Cancer and Chronic Diseases, Faculty of Medicine and Pharmacy of Tangier, Abdelmalek Essaadi University, Tetouan 93000, Morocco;
| | - Majid Omari
- Laboratory of Epidemiology and Research in Health Sciences, Faculty of Medicine, Pharmacy and Dental Medicine, Sidi Mohamed Ben Abdallah University, Fez 30070, Morocco; (M.O.); (K.E.R.); (C.N.)
| | - Oumnia Bouaddi
- Department of Public Health and Clinical Research, Mohammed VI Center for Research and Innovation, Rabat 10112, Morocco; (O.B.); (E.M.E.F.); (S.L.); (S.B.)
- Mohammed VI International School of Public Health, Mohammed VI University of Sciences and Health, Casablanca 82403, Morocco
| | - Elodie Faure
- International Agency for Research on Cancer, World Health Organization, 69366 Lyon, France; (E.F.); (I.H.)
- Center of Epidemiology and Population Health, Inserm, UMR 1018, Paris Saclay University, 94805 Villejuif, France
| | - El Mostafa El Fahime
- Department of Public Health and Clinical Research, Mohammed VI Center for Research and Innovation, Rabat 10112, Morocco; (O.B.); (E.M.E.F.); (S.L.); (S.B.)
| | - Sihame Lkhoyaali
- Department of Public Health and Clinical Research, Mohammed VI Center for Research and Innovation, Rabat 10112, Morocco; (O.B.); (E.M.E.F.); (S.L.); (S.B.)
- Department of Medical Oncology, National Institute of Oncology, Rabat 6213, Morocco
| | - Saber Boutayeb
- Department of Public Health and Clinical Research, Mohammed VI Center for Research and Innovation, Rabat 10112, Morocco; (O.B.); (E.M.E.F.); (S.L.); (S.B.)
- Department of Medical Oncology, National Institute of Oncology, Rabat 6213, Morocco
| | - Karima El Rhazi
- Laboratory of Epidemiology and Research in Health Sciences, Faculty of Medicine, Pharmacy and Dental Medicine, Sidi Mohamed Ben Abdallah University, Fez 30070, Morocco; (M.O.); (K.E.R.); (C.N.)
| | - Chakib Nejjari
- Laboratory of Epidemiology and Research in Health Sciences, Faculty of Medicine, Pharmacy and Dental Medicine, Sidi Mohamed Ben Abdallah University, Fez 30070, Morocco; (M.O.); (K.E.R.); (C.N.)
- Euromed Research Center, Euromed University of Fez, Fez 51, Morocco
| | - Inge Huybrechts
- International Agency for Research on Cancer, World Health Organization, 69366 Lyon, France; (E.F.); (I.H.)
- French Network for Nutrition and Cancer Research (Nacre Network), 78350 Jouy-en-Josas, France
| | - Mohamed Khalis
- Department of Public Health and Clinical Research, Mohammed VI Center for Research and Innovation, Rabat 10112, Morocco; (O.B.); (E.M.E.F.); (S.L.); (S.B.)
- Mohammed VI International School of Public Health, Mohammed VI University of Sciences and Health, Casablanca 82403, Morocco
- International Agency for Research on Cancer, World Health Organization, 69366 Lyon, France; (E.F.); (I.H.)
- Higher Institute of Nursing Professions and Health Techniques, Ministry of Health and Social Protection, Rabat 10000, Morocco
| |
Collapse
|
5
|
Hontecillas-Prieto L, García-Domínguez DJ, Jiménez-Cortegana C, Nogales-Fernández E, Palazón-Carrión N, García-Sancho AM, Ríos-Herranz E, Gumà-Padrò J, Provencio-Pulla M, Rueda-Domínguez A, de la Cruz-Merino L, Sánchez-Margalet V. Obesity and overweight in R/R DLBCL patients is associated with a better response to treatment of R2-GDP-GOTEL trial. Potential role of NK CD8 + cells and vitamin D. Cancer Metab 2025; 13:12. [PMID: 40038834 DOI: 10.1186/s40170-025-00381-7] [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: 11/06/2024] [Accepted: 02/23/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Diffuse large B-cell lymphoma (DLBCL) is the most common type of non-Hodgkin's lymphoma worldwide and is characterized by its heterogeneity. Although first-line therapy improves survival outcomes for DLBCL patients, approximately one third will relapse, often with a poor prognosis. Among the factors influencing prognosis and response to treatment in cancer patients, including those with lymphoma, overweight and obesity have emerged as significant considerations. However, the role of excess weight in DLBCL remains controversial, with studies reporting both negative and positive effects on cancer outcomes. In this translational substudy of the R2-GDP-GOTEL trial, we have evaluated the impact of excess weight as a predictor of treatment response and survival in patients with relapsed/refractory (R/R) DLBCL, and examining its relationship with immune cell dynamics. METHODS Of the 79 patients who received the R2-GDP scheme in the phase II trial, weight and height parameters were obtained in 75 patients before starting treatment. Blood samples were analyzed by flow cytometry. Statistical analyses were performed to determine the prognostic value of overweight and obesity at baseline in R/R DLBCL patients. RESULTS Our results indicate that overweight (including obese) patients exhibit longer survival compared to patients of ideal weight. This group also demonstrated a reduction of regulatory T cells with supposedly protumor activity and an increase of Natural Killer (NK)-like T cells with supposedly antitumor activity. Additionally, we have found that excess weight correlates with better treatment response, associated with elevated levels of vitamin D and CD8 + NK cells. CONCLUSIONS Our findings suggest that excess weight does not exacerbate the progression of DLBCL. Instead, it appears to confer a survival advantage and improve treatment response, with the immune system playing a possible pivotal role in mediating these effects. TRIAL REGISTRATION EudraCT, ID:2014-001620-29.
Collapse
Affiliation(s)
- Lourdes Hontecillas-Prieto
- Clinical Biochemistry Service, Virgen Macarena University Hospital, University of Seville, Seville, Spain
- Department of Medical Biochemistry and Molecular Biology and Immunology, Medical School, Virgen Macarena University Hospital, University of Seville, Seville, Spain
- Clinical Oncology Service, Hospital Universitario Virgen Macarena, University of Seville, Seville, Spain
| | - Daniel J García-Domínguez
- Department of Medical Biochemistry and Molecular Biology and Immunology, Medical School, Virgen Macarena University Hospital, University of Seville, Seville, Spain
- Institute of Biomedicine of Seville, Virgen Macarena University Hospital, CSIC, University of Seville, Seville, Spain
| | - Carlos Jiménez-Cortegana
- Department of Medical Biochemistry and Molecular Biology and Immunology, Medical School, Virgen Macarena University Hospital, University of Seville, Seville, Spain
| | - Esteban Nogales-Fernández
- Clinical Oncology Service, Hospital Universitario Virgen Macarena, University of Seville, Seville, Spain
- Department of Medicine, University of Seville, Seville, Spain
| | - Natalia Palazón-Carrión
- Clinical Oncology Service, Hospital Universitario Virgen Macarena, University of Seville, Seville, Spain
- Department of Medicine, University of Seville, Seville, Spain
| | - Alejandro Martín García-Sancho
- Department of Hematology, Hospital Universitario de Salamanca, IBSAL, CIBERONC, University of Salamanca, Salamanca, Spain
| | | | - Josep Gumà-Padrò
- Department of Clinical Oncology, Hospital Universitari Sant Joan de Reus URV, IISPV, Reus, Spain
| | - Mariano Provencio-Pulla
- Department of Medical Oncology, Facultad de Medicina, Hospital Universitario Puerta de Hierro-Majadahonda, Universidad Autónoma de Madrid, IDIPHISA, Madrid, Spain
| | | | - Luis de la Cruz-Merino
- Clinical Oncology Service, Hospital Universitario Virgen Macarena, University of Seville, Seville, Spain.
- Institute of Biomedicine of Seville, Virgen Macarena University Hospital, CSIC, University of Seville, Seville, Spain.
- Department of Medicine, University of Seville, Seville, Spain.
| | - Víctor Sánchez-Margalet
- Clinical Biochemistry Service, Virgen Macarena University Hospital, University of Seville, Seville, Spain.
- Department of Medical Biochemistry and Molecular Biology and Immunology, Medical School, Virgen Macarena University Hospital, University of Seville, Seville, Spain.
- Institute of Biomedicine of Seville, Virgen Macarena University Hospital, CSIC, University of Seville, Seville, Spain.
| |
Collapse
|
6
|
Zhao C, Zhang T, Xue ST, Zhang P, Wang F, Li Y, Liu Y, Zhao L, Wu J, Yan Y, Mao X, Chen Y, Yuan J, Li Z, Li K. Adipocyte-derived glutathione promotes obesity-related breast cancer by regulating the SCARB2-ARF1-mTORC1 complex. Cell Metab 2025; 37:692-707.e9. [PMID: 39442522 DOI: 10.1016/j.cmet.2024.09.013] [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: 02/23/2024] [Revised: 06/18/2024] [Accepted: 09/24/2024] [Indexed: 10/25/2024]
Abstract
Obesity is a major risk factor for poor breast cancer outcomes, but the impact of obesity-induced tumor microenvironment (TME) metabolites on breast cancer growth and metastasis remains unclear. Here, we performed TME metabolomic analysis in high-fat diet (HFD) mouse models and found that glutathione (GSH) levels were elevated in the TME of obesity-accelerated breast cancer. The deletion of glutamate-cysteine ligase catalytic subunit (GCLC), the rate-limiting enzyme in GSH biosynthesis, in adipocytes but not tumor cells reduced obesity-related tumor progression. Mechanistically, we identified that GSH entered tumor cells and directly bound to lysosomal integral membrane protein-2 (scavenger receptor class B, member 2 [SCARB2]), interfering with the interaction between its N and C termini. This, in turn, recruited mTORC1 to lysosomes through ARF1, leading to the activation of mTOR signaling. Overall, we demonstrated that GSH links obesity and breast cancer progression by acting as an activator of mTOR signaling. Targeting the GSH/SCARB2/mTOR axis could benefit breast cancer patients with obesity.
Collapse
Affiliation(s)
- Chenxi Zhao
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Department of Anesthesiology and Perioperative medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Tingting Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Si-Tu Xue
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Peitao Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Feng Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yunxuan Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Ying Liu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Luyao Zhao
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jie Wu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yechao Yan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xiaoyun Mao
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang 110001, Liaoning, China
| | - Yuping Chen
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Department of Anesthesiology and Perioperative medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China; Cancer Center, Tongji University School of Medicine, Shanghai 200331, China
| | - Jian Yuan
- Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China; Cancer Center, Tongji University School of Medicine, Shanghai 200331, China
| | - Zhuorong Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Ke Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
7
|
Nikita N, Sun Z, Sharma S, Shaver A, Seewaldt V, Lu-Yao G. Epigenetic Landscapes of Aging in Breast Cancer Survivors: Unraveling the Impact of Therapeutic Interventions-A Scoping Review. Cancers (Basel) 2025; 17:866. [PMID: 40075712 PMCID: PMC11899678 DOI: 10.3390/cancers17050866] [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: 01/23/2025] [Revised: 02/24/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Breast cancer therapies have dramatically improved survival rates, but their long-term effects, especially on aging survivors, need careful consideration. This review delves into how breast cancer treatments and aging intersect, focusing on the epigenetic changes triggered by chemotherapy, radiation, hormonal treatments, and targeted therapies. Treatments can speed up biological aging by altering DNA methylation, histone modifications, and chromatin remodeling, affecting gene expression without changing the DNA sequence itself. The review explains the double-edged sword effect of therapy-induced epigenetic modifications, which help fight cancer but also accelerate aging. Chemotherapy and targeted therapies, in particular, impact DNA methylation and histone modifications, promoting chronic inflammation and shortening telomeres. These changes increase biological age, as seen in epigenetic clocks and biomarkers like p21, which also play roles in drug resistance and therapeutic decisions. Chronic inflammation, driven by higher levels of inflammatory cytokines such as TNF-α and IL-6 as well as telomere shortening, significantly contributes to the aging characteristics of breast cancer survivors. Non-coding RNAs, including microRNAs and long non-coding RNAs, are crucial in regulating gene expression and aging pathways altered by these treatments. This review explores new therapies targeting these epigenetic changes, like DNA methylation inhibitors, histone deacetylase inhibitors, and microRNA-based treatments, to reduce the aging effects of cancer therapy. Non-drug approaches, such as dietary changes and lifestyle modifications, also show promise in combating therapy-induced aging. It also highlights the clinical signs of aging-related side effects, such as heart and lung problems, endocrine and reproductive issues, and reduced quality of life. The development of comprehensive methods like the CHEMO-RADIAT score to predict major cardiovascular events after therapy is discussed. Understanding the epigenetic changes caused by breast cancer therapies offers valuable insights for creating interventions to enhance the health span and quality of life for survivors. Continued research is crucial to fully understand these epigenetic alterations and their long-term health impacts.
Collapse
Affiliation(s)
- Nikita Nikita
- Department of Medical Oncology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA; (N.N.); (Z.S.); (S.S.)
- Sidney Kimmel Comprehensive Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Zhengyang Sun
- Department of Medical Oncology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA; (N.N.); (Z.S.); (S.S.)
- Department of Pharmacology, Physiology and Cancer Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Swapnil Sharma
- Department of Medical Oncology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA; (N.N.); (Z.S.); (S.S.)
- Sidney Kimmel Comprehensive Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Amy Shaver
- Department of Medical Oncology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA; (N.N.); (Z.S.); (S.S.)
- Sidney Kimmel Comprehensive Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Victoria Seewaldt
- Department of Population Sciences, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA;
| | - Grace Lu-Yao
- Department of Medical Oncology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA; (N.N.); (Z.S.); (S.S.)
- Sidney Kimmel Comprehensive Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
8
|
Tripathi S, Sharma Y, Kumar D. Unveiling the link between chronic inflammation and cancer. Metabol Open 2025; 25:100347. [PMID: 39876904 PMCID: PMC11772974 DOI: 10.1016/j.metop.2025.100347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 01/05/2025] [Accepted: 01/06/2025] [Indexed: 01/31/2025] Open
Abstract
The highly nuanced transition from an inflammatory process to tumorigenesis is of great scientific interest. While it is well known that environmental stimuli can cause inflammation, less is known about the oncogenic modifications that chronic inflammation in the tissue microenvironment can bring about, as well as how these modifications can set off pro-tumorigenic processes. It is clear that no matter where the environmental factors come from, maintaining an inflammatory microenvironment encourages carcinogenesis. In addition to encouraging angiogenesis and metastatic processes, sustaining the survival and proliferation of malignant transformed cells, and possibly altering the efficacy of therapeutic agents, inflammation can negatively regulate the antitumoral adaptive and innate immune responses. Because chronic inflammation has multiple pathways involved in tumorigenesis and metastasis, it has gained recognition as a marker of cancer and a desirable target for cancer therapy. Recent advances in our knowledge of the molecular mechanisms that drive cancer's progression demonstrate that inflammation promotes tumorigenesis and metastasis while suppressing anti-tumor immunity. In many solid tumor types, including breast, lung, and liver cancer, inflammation stimulates the activation of oncogenes and impairs the body's defenses against the tumor. Additionally, it alters the microenvironment of the tumor. As a tactical approach to cancer treatment, these findings have underscored the importance of targeting inflammatory pathways. This review highlights the role of inflammation in cancer development and metastasis, focusing on its impact on tumor progression, immune suppression, and therapy resistance. It examines current anti-inflammatory strategies, including NSAIDs, cytokine modulators, and STAT3 inhibitors, while addressing their potential and limitations. The review emphasizes the need for further research to unravel the complex mechanisms linking inflammation to cancer progression and identify molecular targets for specific cancer subtypes.
Collapse
Affiliation(s)
- Siddhant Tripathi
- Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra, 411038, India
| | - Yashika Sharma
- Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra, 411038, India
| | - Dileep Kumar
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| |
Collapse
|
9
|
Kim H, Baek S, Han S, Kim GM, Sohn J, Rhee Y, Hong N, Kim MH. Low Skeletal Muscle Radiodensity Predicts Response to CDK4/6 Inhibitors Plus Aromatase Inhibitors in Advanced Breast Cancer. J Cachexia Sarcopenia Muscle 2025; 16:e13666. [PMID: 39686815 DOI: 10.1002/jcsm.13666] [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: 05/24/2024] [Revised: 10/16/2024] [Accepted: 10/31/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Recent evidence indicates that a dysregulated host metabolism influences treatment outcomes in patients with breast cancer. We investigated the association of computed tomography (CT)-derived body composition indices with therapeutic responses in patients with hormone receptor-positive, HER2-negative advanced breast cancer (ABC) on endocrine plus CDK4/6 inhibitor (CDK4/6i) treatment. METHODS The study involved a retrospective cohort of patients with ABC at the Yonsei Cancer Center who received CDK4/6i and aromatase inhibitors as first-line therapy between January 2017 and October 2020. Body composition parameters were estimated from the non-enhanced CT images of the third lumbar spine by commercialized deep learning software. Patients with low skeletal muscle radiodensity (SMD) were defined as patients with SMD of low tertile (≤ 28.7 Hounsfield Units). The primary outcome was progression-free survival (PFS). RESULTS Among the 247 female participants (median age, 53 years; mean body mass index [BMI], 23.7 kg/m2), 45.7% had disease progression or death during a median follow-up of 36.4 months. After adjusting for age and visceral metastasis, SMD was the only independent predictor among body composition parameters for worse PFS (adjusted hazard ratio [HR] = 1.20 per standard deviation decrement, 95% CI: 1.01-1.42, p = 0.041), whereas BMI, muscle area, and fat area were not. Participants with low SMD had a higher risk of progression than those without (PFS, 27.2 vs. 51.1 months, p = 0.009; adjusted HR 1.84, 95% CI: 1.22-2.76, p = 0.003). Strong associations between low SMD and poor PFS were observed in groups with pre-menopause status (HR, 3.04 vs. 1.19 in post-menopause; 95% CI: 1.54-5.99, p for interaction < 0.05) and without visceral metastases (HR, 2.95 vs. 1.19 in with visceral metastases; 95% CI: 1.59-5.49, p for interaction < 0.05). CONCLUSIONS CT-defined low SMD predicts poor treatment outcomes in patients with ABC undergoing first-line treatment with aromatase inhibitors and CDK4/6i.
Collapse
Affiliation(s)
- Hyunwook Kim
- Department of Internal Medicine, Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Seungjin Baek
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Sookyeong Han
- Endocrine Research Institute, Severance Hospital, Seoul, South Korea
| | - Gun Min Kim
- Department of Internal Medicine, Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Joohyuk Sohn
- Department of Internal Medicine, Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Yumie Rhee
- Department of Internal Medicine, Endocrine Research Institute, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Namki Hong
- Department of Internal Medicine, Endocrine Research Institute, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Min Hwan Kim
- Department of Internal Medicine, Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
10
|
Torabinejad S, Miro C, Nappi A, Del Giudice F, Cicatiello AG, Sagliocchi S, Acampora L, Restolfer F, Murolo M, Di Cicco E, Capone F, Imbimbo C, Dentice M, Crocetto F. Obesity alters the fitness of peritumoral adipose tissue, exacerbating tumor invasiveness in renal cancer through the induction of ADAM12 and CYP1B1. Mol Oncol 2025. [PMID: 39806854 DOI: 10.1002/1878-0261.13782] [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: 07/08/2024] [Revised: 10/21/2024] [Accepted: 11/28/2024] [Indexed: 01/16/2025] Open
Abstract
Obesity exacerbates the risk and aggressiveness of many types of cancer. Adipose tissue (AT) represents a prevalent component of the tumor microenvironment (TME) and contributes to cancer development and progression. Reciprocal communication between cancer and adipose cells leads to the generation of cancer-associated adipocytes (CAAs), which in turn foster tumor invasiveness by producing paracrine metabolites, adipocytokines, and growth factors. Interfering with the crosstalk between CAAs and cancer cells is of key relevance in the prevention of tumor progression. The present study aimed to analyze the contribution of peritumoral AT in renal cell carcinoma (RCC) progression in lean versus overweight or obese patients. By isolating human adipose-derived stromal/stem cells from the three groups of patients and performing conditioned medium studies with RCC cells along with in vivo xenograft experiments, we found that peritumoral adipocytes from the three groups show a distinct expression profile of genes. In particular, ADAM metallopeptidase domain 12 (ADAM12) and cytochrome P450 family 1 subfamily B member 1 (CYP1B1) were found to be upregulated in obesity and their silencing reduced RCC cell invasiveness. In conclusion, high ADAM12 and CYP1B1 expressions in the peritumoral adipocytes boost tumor invasiveness and may serve as an indicator of poor prognosis in RCC.
Collapse
Affiliation(s)
- Sepehr Torabinejad
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Italy
| | - Caterina Miro
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Italy
| | - Annarita Nappi
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Italy
| | - Francesco Del Giudice
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", Italy
| | | | - Serena Sagliocchi
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Italy
| | - Lucia Acampora
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Italy
| | - Federica Restolfer
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Italy
| | - Melania Murolo
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Italy
| | - Emery Di Cicco
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Italy
| | - Federico Capone
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", Italy
| | - Ciro Imbimbo
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", Italy
| | - Monica Dentice
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Italy
- CEINGE - Biotecnologie Avanzate S.c.a.r.l., Naples, Italy
| | - Felice Crocetto
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", Italy
| |
Collapse
|
11
|
Jiménez-Cortegana C, Sánchez-Jiménez F, De La Cruz-Merino L, Sánchez-Margalet V. Role of Sam68 in different types of cancer (Review). Int J Mol Med 2025; 55:3. [PMID: 39450529 PMCID: PMC11537268 DOI: 10.3892/ijmm.2024.5444] [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: 07/08/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
Src‑associated in mitosis 68 kDa protein (Sam68) is a protein encoded by the heteronuclear ribonucleoprotein particle K homology (KH) single domain‑containing, RNA‑binding, signal transduction‑associated protein 1 (known as KHDRBS1) gene in humans. This protein contains binding sites for critical components in a variety of cellular processes, including the regulation of gene expression, RNA processing and cell signaling. Thus, Sam68 may play a role in a variety of diseases, including cancer. Sam68 has been widely demonstrated to participate in tumor cell proliferation, progression and metastasis to be involved in the regulation of cancer stem cell self‑renewal. Based on the body of evidence available, Sam68 emerges as a promising target for this disease. The objectives of the present included summarizing the role of Sam68 in cancer murine models and cancer patients, unraveling the molecular mechanisms underlying its oncogenic potential and discussing the effectiveness of antitumor agents in reducing the malignant effects of Sam68 during tumorigenesis.
Collapse
Affiliation(s)
- Carlos Jiménez-Cortegana
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Flora Sánchez-Jiménez
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, 41009 Seville, Spain
- Department of Laboratory Medicine, Virgen Macarena University Hospital, 41009 Seville, Spain
| | - Luis De La Cruz-Merino
- Department of Medicine, School of Medicine, University of Seville, 41009 Seville, Spain
- Medical Oncology Service, Virgen Macarena University Hospital, 41009 Seville, Spain
- Institute of Biomedicine of Seville, Virgen Macarena University Hospital, Consejo Superior de Investigaciones Científicas, University of Seville, 41013 Seville, Spain
| | - Víctor Sánchez-Margalet
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, 41009 Seville, Spain
- Department of Laboratory Medicine, Virgen Macarena University Hospital, 41009 Seville, Spain
- Institute of Biomedicine of Seville, Virgen Macarena University Hospital, Consejo Superior de Investigaciones Científicas, University of Seville, 41013 Seville, Spain
| |
Collapse
|
12
|
Calabrese C, Miserocchi G, De Vita A, Spadazzi C, Cocchi C, Vanni S, Gabellone S, Martinelli G, Ranallo N, Bongiovanni A, Liverani C. Lipids and adipocytes involvement in tumor progression with a focus on obesity and diet. Obes Rev 2024; 25:e13833. [PMID: 39289899 DOI: 10.1111/obr.13833] [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: 10/23/2023] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 09/19/2024]
Abstract
The adipose tissue is a complex organ that can play endocrine, metabolic, and immune regulatory roles in cancer. In particular, adipocytes provide metabolic substrates for cancer cell proliferation and produce signaling molecules that can stimulate cell adhesion, migration, invasion, angiogenesis, and inflammation. Cancer cells, in turn, can reprogram adipocytes towards a more inflammatory state, resulting in a vicious cycle that fuels tumor growth and evolution. These mechanisms are enhanced in obesity, which is associated with the risk of developing certain tumors. Diet, an exogenous source of lipids with pro- or anti-inflammatory functions, has also been connected to cancer risk. This review analyzes how adipocytes and lipids are involved in tumor development and progression, focusing on the relationship between obesity and cancer. In addition, we discuss how diets with varying lipid intakes can affect the disease outcomes. Finally, we introduce novel metabolism-targeted treatments and adipocyte-based therapies in oncology.
Collapse
Affiliation(s)
- Chiara Calabrese
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Giacomo Miserocchi
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Alessandro De Vita
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Chiara Spadazzi
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Claudia Cocchi
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Silvia Vanni
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Sofia Gabellone
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Giovanni Martinelli
- Scientific Directorate, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Nicoletta Ranallo
- Clinical and Experimental Oncology, Immunotherapy, Rare Cancers and Biological Resource Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Alberto Bongiovanni
- Clinical and Experimental Oncology, Immunotherapy, Rare Cancers and Biological Resource Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Chiara Liverani
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| |
Collapse
|
13
|
Yum C, Andolino C, Layosa MA, Coleman M, Hursting SD, Teegarden D. Differential effects of leptin on energy metabolism in murine cell models of metastatic triple negative breast cancer. Diabetol Metab Syndr 2024; 16:288. [PMID: 39609706 PMCID: PMC11603625 DOI: 10.1186/s13098-024-01535-1] [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: 08/29/2024] [Accepted: 11/17/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND Leptin, an energy balance regulator secreted by adipocytes, increases metastatic potential of breast cancer cells. The impact on cancer cell metabolism remains unclear given that most studies of leptin and breast cancer cell metabolism utilize supraphysiological glucose concentrations. METHODS Using two murine models of metastatic triple-negative breast cancer (TNBC) differing in genetic alterations (4T1: p53 and Pik3ca mutations; metM-Wntlung: increased Wnt signaling) and cultured in physiological (5 mM) glucose media, we tested the hypothesis that leptin increases migration of metastatic breast cancer cells through regulation of glucose metabolism. RESULTS Our results showed that leptin treatment, compared with vehicle, increased cell migration in each cell line, with decreased leptin receptor (Ob-R) mRNA expression in 4T1, but not metM-Wntlung, cells. AMP-activated protein kinase (AMPK) was activated in 4T1 with leptin treatment but decreased in metM-Wntlung. Leptin decreased fatty acid synthase (Fasn) and carnitine palmitoyltransferase 1a (Cpt1a) mRNA expression in 4T1 cells but increased their expression in metM-Wntlung cells. Fatty acid oxidation was not necessary for leptin-induced migration in either cell line. Leptin increased palmitate synthesis from glucose in metM-Wntlung, but not 4T1 cells. Moreover, although leptin increased glucose transporter 1 (Glut1) mRNA expression in both cell lines and inhibition of glycolysis blocked leptin-induced migration in metM-Wntlung, but not 4T1 cells. CONCLUSION Taken together, these results demonstrate that at physiological glucose concentrations, leptin increases migration of 4T1 and metM-Wntlung cells via shared and distinct effects on energy metabolism, suggesting that the type of TNBC genetic alteration plays a role in differential metabolic regulation of leptin-induced migration.
Collapse
Affiliation(s)
- Chaehyun Yum
- Department of Nutrition Science, Interdepartmental Nutrition Program, Purdue University, West Lafayette, IN, 47907, USA
| | - Chaylen Andolino
- Department of Nutrition Science, Interdepartmental Nutrition Program, Purdue University, West Lafayette, IN, 47907, USA
- Purdue University Institute for Cancer Research, Purdue University, West Lafayette, IN, 47907, USA
| | - Marjorie Anne Layosa
- Department of Nutrition Science, Interdepartmental Nutrition Program, Purdue University, West Lafayette, IN, 47907, USA
- Purdue University Institute for Cancer Research, Purdue University, West Lafayette, IN, 47907, USA
| | - Michael Coleman
- Department of Nutrition, University of North Carolina, Chapel Hill, NC, 27516, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27516, USA
| | - Stephen D Hursting
- Department of Nutrition, University of North Carolina, Chapel Hill, NC, 27516, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27516, USA
| | - Dorothy Teegarden
- Department of Nutrition Science, Interdepartmental Nutrition Program, Purdue University, West Lafayette, IN, 47907, USA.
- Purdue University Institute for Cancer Research, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
14
|
Lagarde CB, Thapa K, Cullen NM, Hawes ML, Salim K, Benz MC, Dietrich SR, Burow BE, Bunnell BA, Martin EC, Collins-Burow BM, Lynch RM, Hoang VT, Burow ME, Fang JS. Obesity and leptin in breast cancer angiogenesis. Front Endocrinol (Lausanne) 2024; 15:1465727. [PMID: 39439572 PMCID: PMC11493622 DOI: 10.3389/fendo.2024.1465727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 09/04/2024] [Indexed: 10/25/2024] Open
Abstract
At the time of breast cancer diagnosis, most patients meet the diagnostic criteria to be classified as obese or overweight. This can significantly impact patient outcome: breast cancer patients with obesity (body mass index > 30) have a poorer prognosis compared to patients with a lean BMI. Obesity is associated with hyperleptinemia, and leptin is a well-established driver of metastasis in breast cancer. However, the effect of hyperleptinemia on angiogenesis in breast cancer is less well-known. Angiogenesis is an important process in breast cancer because it is essential for tumor growth beyond 1mm3 in size as well as cancer cell circulation and metastasis. This review investigates the role of leptin in regulating angiogenesis, specifically within the context of breast cancer and the associated tumor microenvironment in obese patients.
Collapse
Affiliation(s)
- Courtney B. Lagarde
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, United States
- Tulane University Cancer Center, New Orleans, LA, United States
| | - Kapil Thapa
- Department of Cell and Molecular Biology, Tulane University School of Science and Engineering, New Orleans, LA, United States
| | - Nicole M. Cullen
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, United States
- Tulane University Cancer Center, New Orleans, LA, United States
| | - Mackenzie L. Hawes
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, United States
- Tulane University Cancer Center, New Orleans, LA, United States
| | - Khudeja Salim
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, United States
- Tulane University Cancer Center, New Orleans, LA, United States
| | - Megan C. Benz
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, United States
- Tulane University Cancer Center, New Orleans, LA, United States
| | - Sophie R. Dietrich
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, United States
- Tulane University Cancer Center, New Orleans, LA, United States
- United States Department of Agriculture Southern Regional Research Center, New Orleans, LA, United States
| | - Brandon E. Burow
- Department of Cell and Molecular Biology, Tulane University School of Science and Engineering, New Orleans, LA, United States
| | - Bruce A. Bunnell
- Department of Microbiology, Immunology, and Genetics, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Elizabeth C. Martin
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, United States
- Tulane University Cancer Center, New Orleans, LA, United States
| | - Bridgette M. Collins-Burow
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, United States
- Tulane University Cancer Center, New Orleans, LA, United States
| | - Ronald M. Lynch
- Department of Physiology, College of Medicine, University of Arizona, Tucson, AZ, United States
| | - Van T. Hoang
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, United States
- Tulane University Cancer Center, New Orleans, LA, United States
| | - Matthew E. Burow
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, LA, United States
- Tulane University Cancer Center, New Orleans, LA, United States
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, United States
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, United States
| | - Jennifer S. Fang
- Department of Cell and Molecular Biology, Tulane University School of Science and Engineering, New Orleans, LA, United States
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA, United States
| |
Collapse
|
15
|
Bhattacharya P, Linnenbach A, South AP, Martinez-Outschoorn U, Curry JM, Johnson JM, Harshyne LA, Mahoney MG, Luginbuhl AJ, Vadigepalli R. Tumor microenvironment governs the prognostic landscape of immunotherapy for head and neck squamous cell carcinoma: A computational model-guided analysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.26.615149. [PMID: 39386511 PMCID: PMC11463398 DOI: 10.1101/2024.09.26.615149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Immune checkpoint inhibition (ICI) has emerged as a critical treatment strategy for squamous cell carcinoma of the head and neck (HNSCC) that halts the immune escape of the tumor cells. Increasing evidence suggests that the onset, progression, and lack of/no response of HNSCC to ICI are emergent properties arising from the interactions within the tumor microenvironment (TME). Deciphering how the diversity of cellular and molecular interactions leads to distinct HNSCC TME subtypes subsequently governing the ICI response remains largely unexplored. We developed a cellular-molecular model of the HNSCC TME that incorporates multiple cell types, cellular states, and transitions, and molecularly mediated paracrine interactions. An exhaustive simulation of the HNSCC TME network shows that distinct mechanistic balances within the TME give rise to the five clinically observed TME subtypes such as immune/non-fibrotic, immune/fibrotic, fibrotic only and immune/fibrotic desert. We predict that the cancer-associated fibroblast, beyond a critical proliferation rate, drastically worsens the ICI response by hampering the accessibility of the CD8+ killer T cells to the tumor cells. Our analysis reveals that while an Interleukin-2 (IL-2) + ICI combination therapy may improve response in the immune desert scenario, Osteopontin (OPN) and Leukemia Inhibition Factor (LIF) knockout with ICI yields the best response in a fibro-dominated scenario. Further, we predict Interleukin-8 (IL-8), and lactate can serve as crucial biomarkers for ICI-resistant HNSCC phenotypes. Overall, we provide an integrated quantitative framework that explains a wide range of TME-mediated resistance mechanisms for HNSCC and predicts TME subtype-specific targets that can lead to an improved ICI outcome.
Collapse
Affiliation(s)
- Priyan Bhattacharya
- Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA19107, USA
| | - Alban Linnenbach
- Department of Otolaryngology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA19107, USA
| | - Andrew P. South
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA19107, USA
| | - Ubaldo Martinez-Outschoorn
- Department of Medical Oncology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA19107, USA
| | - Joseph M. Curry
- Department of Otolaryngology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA19107, USA
| | - Jennifer M. Johnson
- Department of Otolaryngology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA19107, USA
- Department of Medical Oncology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA19107, USA
| | - Larry A. Harshyne
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA19107, USA
| | - Mỹ G. Mahoney
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA19107, USA
| | - Adam J. Luginbuhl
- Department of Otolaryngology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA19107, USA
| | - Rajanikanth Vadigepalli
- Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA19107, USA
| |
Collapse
|
16
|
Sergi D, Melloni M, Passaro A, Neri LM. Influence of Type 2 Diabetes and Adipose Tissue Dysfunction on Breast Cancer and Potential Benefits from Nutraceuticals Inducible in Microalgae. Nutrients 2024; 16:3243. [PMID: 39408212 PMCID: PMC11478231 DOI: 10.3390/nu16193243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Breast cancer (BC) represents the most prevalent cancer in women at any age after puberty. From a pathogenetic prospective, despite a wide array of risk factors being identified thus far, poor metabolic health is emerging as a putative risk factor for BC. In particular, type 2 diabetes mellitus (T2DM) provides a perfect example bridging the gap between poor metabolic health and BC risk. Indeed, T2DM is preceded by a status of hyperinsulinemia and is characterised by hyperglycaemia, with both factors representing potential contributors to BC onset and progression. Additionally, the aberrant secretome of the dysfunctional, hypertrophic adipocytes, typical of obesity, characterised by pro-inflammatory mediators, is a shared pathogenetic factor between T2DM and BC. In this review, we provide an overview on the effects of hyperglycaemia and hyperinsulinemia, hallmarks of type 2 diabetes mellitus, on breast cancer risk, progression, treatment and prognosis. Furthermore, we dissect the role of the adipose-tissue-secreted adipokines as additional players in the pathogenesis of BC. Finally, we focus on microalgae as a novel superfood and a source of nutraceuticals able to mitigate BC risk by improving metabolic health and targeting cellular pathways, which are disrupted in the context of T2DM and obesity.
Collapse
Affiliation(s)
- Domenico Sergi
- Department of Translational Medicine, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy; (D.S.); (M.M.)
| | - Mattia Melloni
- Department of Translational Medicine, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy; (D.S.); (M.M.)
| | - Angelina Passaro
- Department of Translational Medicine, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy; (D.S.); (M.M.)
| | - Luca Maria Neri
- Department of Translational Medicine, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy; (D.S.); (M.M.)
- Laboratory for Technologies of Advanced Therapies (LTTA)—Electron Microscopy Center, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy
| |
Collapse
|
17
|
Asiri A, Al Qarni A, Bakillah A. The Interlinking Metabolic Association between Type 2 Diabetes Mellitus and Cancer: Molecular Mechanisms and Therapeutic Insights. Diagnostics (Basel) 2024; 14:2132. [PMID: 39410536 PMCID: PMC11475808 DOI: 10.3390/diagnostics14192132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 09/15/2024] [Accepted: 09/16/2024] [Indexed: 10/20/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) and cancer share common risk factors including obesity, inflammation, hyperglycemia, and hyperinsulinemia. High insulin levels activate the PI3K/Akt/mTOR signaling pathway promoting cancer cell growth, survival, proliferation, metastasis, and anti-apoptosis. The inhibition of the PI3K/Akt/mTOR signaling pathway for cancer remains a promising therapy; however, drug resistance poses a major problem in clinical settings resulting in limited efficacy of agents; thus, combination treatments with therapeutic inhibitors may solve the resistance to such agents. Understanding the metabolic link between diabetes and cancer can assist in improving the therapeutic strategies used for the management of cancer patients with diabetes and vice versa. This review provides an overview of shared molecular mechanisms between diabetes and cancer as well as discusses established and emerging therapeutic anti-cancer agents targeting the PI3K/Akt/mTOR pathway in cancer management.
Collapse
Affiliation(s)
- Abutaleb Asiri
- King Abdullah International Medical Research Center (KAIMRC), Eastern Region, Al Ahsa 36428, Saudi Arabia; (A.A.); (A.A.Q.)
- Division of Medical Research Core-A, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Al Ahsa 36428, Saudi Arabia
- King Abdulaziz Hospital, Ministry of National Guard-Health Affairs (MNG-HA), Al Ahsa 36428, Saudi Arabia
| | - Ali Al Qarni
- King Abdullah International Medical Research Center (KAIMRC), Eastern Region, Al Ahsa 36428, Saudi Arabia; (A.A.); (A.A.Q.)
- Division of Medical Research Core-A, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Al Ahsa 36428, Saudi Arabia
- King Abdulaziz Hospital, Ministry of National Guard-Health Affairs (MNG-HA), Al Ahsa 36428, Saudi Arabia
| | - Ahmed Bakillah
- King Abdullah International Medical Research Center (KAIMRC), Eastern Region, Al Ahsa 36428, Saudi Arabia; (A.A.); (A.A.Q.)
- Division of Medical Research Core-A, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Al Ahsa 36428, Saudi Arabia
- King Abdulaziz Hospital, Ministry of National Guard-Health Affairs (MNG-HA), Al Ahsa 36428, Saudi Arabia
| |
Collapse
|
18
|
Zhang H, Goedegebuure SP, Ding L, DeNardo D, Fields RC, Chen Y, Payne P, Li F. M3NetFlow: A novel multi-scale multi-hop graph AI model for integrative multi-omic data analysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.15.545130. [PMID: 39282437 PMCID: PMC11398409 DOI: 10.1101/2023.06.15.545130] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Multi-omic data-driven studies, characterizing complex disease signaling system from multiple levels, are at the forefront of precision medicine and healthcare. The integration and interpretation of multi-omic data are essential for identifying molecular targets and deciphering core signaling pathways of complex diseases. However, it remains an open problem due the large number of biomarkers and complex interactions among them. In this study, we propose a novel Multi-scale Multi-hop Multi-omic graph model, M3NetFlow, to facilitate generic multi-omic data analysis to rank targets and infer core signaling flows/pathways. To evaluate M3NetFlow, we applied it in two independent multi-omic case studies: 1) uncovering mechanisms of synergistic drug combination response (defined as anchor-target guided learning), and 2) identifying biomarkers and pathways of Alzheimer 's disease (AD). The evaluation and comparison results showed M3NetFlow achieves the best prediction accuracy (accurate), and identifies a set of essential targets and core signaling pathways (interpretable). The model can be directly applied to other multi-omic data-driven studies. The code is publicly accessible at: https://github.com/FuhaiLiAiLab/M3NetFlow.
Collapse
Affiliation(s)
- Heming Zhang
- Institute for Informatics, Data Science and Biostatistics (I2DB), Washington University School of Medicine, St. Louis, MO, USA
| | - S. Peter Goedegebuure
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Li Ding
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - David DeNardo
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Ryan C. Fields
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Yixin Chen
- Department of Computer Science and Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Philip Payne
- Institute for Informatics, Data Science and Biostatistics (I2DB), Washington University School of Medicine, St. Louis, MO, USA
| | - Fuhai Li
- Institute for Informatics, Data Science and Biostatistics (I2DB), Washington University School of Medicine, St. Louis, MO, USA
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
19
|
Yende AS, Sharma D. Obesity, dysbiosis and inflammation: interactions that modulate the efficacy of immunotherapy. Front Immunol 2024; 15:1444589. [PMID: 39253073 PMCID: PMC11381382 DOI: 10.3389/fimmu.2024.1444589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 08/06/2024] [Indexed: 09/11/2024] Open
Abstract
Recent years have seen an outstanding growth in the understanding of connections between diet-induced obesity, dysbiosis and alterations in the tumor microenvironment. Now we appreciate that gut dysbiosis can exert important effects in distant target tissues via specific microbes and metabolites. Multiple studies have examined how diet-induced obese state is associated with gut dysbiosis and how gut microbes direct various physiological processes that help maintain obese state in a bidirectional crosstalk. Another tightly linked factor is sustained low grade inflammation in tumor microenvironment that is modulated by both obese state and dysbiosis, and influences tumor growth as well as response to immunotherapy. Our review brings together these important aspects and explores their connections. In this review, we discuss how obese state modulates various components of the breast tumor microenvironment and gut microbiota to achieve sustained low-grade inflammation. We explore the crosstalk between different components of tumor microenvironment and microbes, and how they might modulate the response to immunotherapy. Discussing studies from multiple tumor types, we delve to find common microbial characteristics that may positively or negatively influence immunotherapy efficacy in breast cancer and may guide future studies.
Collapse
Affiliation(s)
- Ashutosh S Yende
- Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, United States
| | - Dipali Sharma
- Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, United States
| |
Collapse
|
20
|
Frąk M, Grenda A, Krawczyk P, Kuźnar-Kamińska B, Pazdrowski P, Kędra K, Chmielewska I, Milanowski J. The influence of nutritional status, lipid profile, leptin concentration and polymorphism of genes encoding leptin and neuropeptide Y on the effectiveness of immunotherapy in advanced NSCLC patients. BMC Cancer 2024; 24:937. [PMID: 39090596 PMCID: PMC11295594 DOI: 10.1186/s12885-024-12716-6] [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: 06/06/2024] [Accepted: 07/26/2024] [Indexed: 08/04/2024] Open
Abstract
INTRODUCTION Neuropeptide Y is a neurotransmitter in the nervous system and belongs to the orexigenic system that increases appetite. Its excessive secretion leads to obesity. Leptin is a pro-inflammatory adipokine (produced in adipose tissue) induced in obesity and may mediate increased antitumor immunity in obesity (including the promotion of M1 macrophages). Leptin and neuropeptide Y gene polymorphisms, causing increased leptin levels and the occurrence of obesity, and lipid profile disorders, may increase the effectiveness of immunotherapy. MATERIALS AND METHODS In 121 patients with advanced NSCLC without mutations in the EGFR gene and rearrangements of the ALK and ROS1 genes, undergoing immunotherapy (1st and 2nd line of treatment) or chemoimmunotherapy (1st line of treatment), we assessed BMI, lipid profile, PD-L1 expression on cancer cells using the immunohistochemical method (clone SP263 antibody), leptin concentration in blood serum by ELISA, polymorphisms in the promoter region of the genes for leptin (LEP) and neuropeptide Y (NPY) by real-time PCR. RESULTS Leptin concentration was significantly higher in obese patients than in patients with normal or low weight (p = 0.00003) and in patients with disease stabilization compared to patients with progression observed during immunotherapy (p = 0.012). Disease control occurred significantly more often in patients with the GA or AA genotype than patients with the GG genotype in the rs779039 polymorphism of the LEP gene. The median PFS in the entire study group was five months (95% CI: 3-5.5), and the median OS was 12 months (95% CI: 8-16). Median PFS was highest in patients with TPS ≥ 50% (6.5 months) and in obese patients (6.6 months). Obese patients also had a slightly longer median OS compared to other patients (23.8 vs. 13 months). The multivariate Cox logistic regression test showed that the only factor reducing the risk of progression was TPS ≥ 50% (HR = 0.6068, 95% CI: 0.4001-0.9204, p = 0, 0187), and the only factor reducing the risk of death was high leptin concentration (HR = 0.6743, 95% CI: 0.4243-1.0715, p = 0.0953). CONCLUSION Assessment of nutritional status, serum leptin concentration and polymorphisms in the LEP gene may be of additional importance in predicting the effectiveness of immunotherapy and chemoimmunotherapy in patients with advanced NSCLC.
Collapse
Affiliation(s)
- Małgorzata Frąk
- Department of Pneumonology, Oncology and Allergology Medical, University of Lublin, Jaczewskiego 8, Lublin, 20-954, Poland.
| | - Anna Grenda
- Department of Pneumonology, Oncology and Allergology Medical, University of Lublin, Jaczewskiego 8, Lublin, 20-954, Poland.
| | - Paweł Krawczyk
- Department of Pneumonology, Oncology and Allergology Medical, University of Lublin, Jaczewskiego 8, Lublin, 20-954, Poland
| | - Barbara Kuźnar-Kamińska
- Department of Pulmonology, Allergology and Pulmonary Oncology, Poznan University of Medical Sciences, Poznań, Poland
| | - Paweł Pazdrowski
- Department of Head, Neck Surgery and Laryngological Oncology, Poznan University of Medical Sciences, Poznań, Poland
| | - Karolina Kędra
- Institute of Physical Chemistry, Polish Academy of Sciences in Warsaw, Warsaw, Poland
| | - Izabela Chmielewska
- Department of Pneumonology, Oncology and Allergology Medical, University of Lublin, Jaczewskiego 8, Lublin, 20-954, Poland
| | - Janusz Milanowski
- Department of Pneumonology, Oncology and Allergology Medical, University of Lublin, Jaczewskiego 8, Lublin, 20-954, Poland
| |
Collapse
|
21
|
Jiménez-Cortegana C, Gutiérrez-García C, Sánchez-Jiménez F, Vilariño-García T, Flores-Campos R, Pérez-Pérez A, Garnacho C, Sánchez-León ML, García-Domínguez DJ, Hontecillas-Prieto L, Palazón-Carrión N, De La Cruz-Merino L, Sánchez-Margalet V. Impact of obesity‑associated myeloid‑derived suppressor cells on cancer risk and progression (Review). Int J Oncol 2024; 65:79. [PMID: 38940351 PMCID: PMC11251741 DOI: 10.3892/ijo.2024.5667] [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: 04/09/2024] [Accepted: 06/12/2024] [Indexed: 06/29/2024] Open
Abstract
Obesity is a chronic disease caused by the accumulation of excessive adipose tissue. This disorder is characterized by chronic low‑grade inflammation, which promotes the release of proinflammatory mediators, including cytokines, chemokines and leptin. Simultaneously, chronic inflammation can predispose to cancer development, progression and metastasis. Proinflammatory molecules are involved in the recruitment of specific cell populations in the tumor microenvironment. These cell populations include myeloid‑derived suppressor cells (MDSCs), a heterogeneous, immature myeloid population with immunosuppressive abilities. Obesity‑associated MDSCs have been linked with tumor dissemination, progression and poor clinical outcomes. A comprehensive literature review was conducted to assess the impact of obesity‑associated MDSCs on cancer in both preclinical models and oncological patients with obesity. A secondary objective was to examine the key role that leptin, the most important proinflammatory mediator released by adipocytes, plays in MDSC‑driven immunosuppression Finally, an overview is provided of the different therapeutic approaches available to target MDSCs in the context of obesity‑related cancer.
Collapse
Affiliation(s)
- Carlos Jiménez-Cortegana
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Cristian Gutiérrez-García
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Flora Sánchez-Jiménez
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Teresa Vilariño-García
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Rocio Flores-Campos
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Antonio Pérez-Pérez
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Carmen Garnacho
- Department of Normal and Pathological Histology and Cytology, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Maria L. Sánchez-León
- Oncology Service, Virgen Macarena University Hospital, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Daniel J. García-Domínguez
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Lourdes Hontecillas-Prieto
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Natalia Palazón-Carrión
- Oncology Service, Virgen Macarena University Hospital, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Luis De La Cruz-Merino
- Oncology Service, Virgen Macarena University Hospital, School of Medicine, University of Seville, 41009 Seville, Spain
- Institute of Biomedicine of Seville, Virgen Macarena University Hospital, CSIC, University of Seville, Seville 41013, Spain
| | - Víctor Sánchez-Margalet
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
- Institute of Biomedicine of Seville, Virgen Macarena University Hospital, CSIC, University of Seville, Seville 41013, Spain
| |
Collapse
|
22
|
Khalifa A, Guijarro A, Nencioni A. Advances in Diet and Physical Activity in Breast Cancer Prevention and Treatment. Nutrients 2024; 16:2262. [PMID: 39064705 PMCID: PMC11279876 DOI: 10.3390/nu16142262] [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: 05/27/2024] [Revised: 07/07/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
There is currently a growing interest in diets and physical activity patterns that may be beneficial in preventing and treating breast cancer (BC). Mounting evidence indicates that indeed, the so-called Mediterranean diet (MedDiet) and regular physical activity likely both help reduce the risk of developing BC. For those who have already received a BC diagnosis, these interventions may decrease the risk of tumor recurrence after treatment and improve quality of life. Studies also show the potential of other dietary interventions, including fasting or modified fasting, calorie restriction, ketogenic diets, and vegan or plant-based diets, to enhance the efficacy of BC therapies. In this review article, we discuss the biological rationale for utilizing these dietary interventions and physical activity in BC prevention and treatment. We highlight published and ongoing clinical studies that have applied these lifestyle interventions to BC patients. This review offers valuable insights into the potential application of these dietary interventions and physical activity as complimentary therapies in BC management.
Collapse
Affiliation(s)
- Amr Khalifa
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132 Genoa, Italy;
| | - Ana Guijarro
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132 Genoa, Italy;
| | - Alessio Nencioni
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, 16132 Genoa, Italy;
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| |
Collapse
|
23
|
Kakkat S, Suman P, Turbat- Herrera EA, Singh S, Chakroborty D, Sarkar C. Exploring the multifaceted role of obesity in breast cancer progression. Front Cell Dev Biol 2024; 12:1408844. [PMID: 39040042 PMCID: PMC11260727 DOI: 10.3389/fcell.2024.1408844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 06/17/2024] [Indexed: 07/24/2024] Open
Abstract
Obesity is a multifaceted metabolic disorder characterized by excessive accumulation of adipose tissue. It is a well-established risk factor for the development and progression of breast cancer. Adipose tissue, which was once regarded solely as a passive energy storage depot, is now acknowledged as an active endocrine organ producing a plethora of bioactive molecules known as adipokines that contribute to the elevation of proinflammatory cytokines and estrogen production due to enhanced aromatase activity. In the context of breast cancer, the crosstalk between adipocytes and cancer cells within the adipose microenvironment exerts profound effects on tumor initiation, progression, and therapeutic resistance. Moreover, adipocytes can engage in direct interactions with breast cancer cells through physical contact and paracrine signaling, thereby facilitating cancer cell survival and invasion. This review endeavors to summarize the current understanding of the intricate interplay between adipocyte-associated factors and breast cancer progression. Furthermore, by discussing the different aspects of breast cancer that can be adversely affected by obesity, this review aims to shed light on potential avenues for new and novel therapeutic interventions.
Collapse
Affiliation(s)
- Sooraj Kakkat
- Department of Pathology, University of South Alabama, Mobile, AL, United States
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, United States
| | - Prabhat Suman
- Department of Pathology, University of South Alabama, Mobile, AL, United States
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, United States
| | - Elba A. Turbat- Herrera
- Department of Pathology, University of South Alabama, Mobile, AL, United States
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, United States
| | - Seema Singh
- Department of Pathology, University of South Alabama, Mobile, AL, United States
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, United States
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, United States
| | - Debanjan Chakroborty
- Department of Pathology, University of South Alabama, Mobile, AL, United States
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, United States
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, United States
| | - Chandrani Sarkar
- Department of Pathology, University of South Alabama, Mobile, AL, United States
- Cancer Biology Program, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, United States
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, United States
| |
Collapse
|
24
|
Marcarini JAC, Grippa WR, Neto LCBS, Podestá OPG, Bolsoni-Lopes A, Nunes KZ, Lopes-Júnior LC. Nutritional status of women with non-metastatic breast cancer receiving outpatient chemotherapy. Nutrition 2024; 123:112411. [PMID: 38518541 DOI: 10.1016/j.nut.2024.112411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 02/14/2024] [Accepted: 02/24/2024] [Indexed: 03/24/2024]
Abstract
OBJECTIVES The aim of this study was to evaluate and compare the nutritional status of women with stage I to III breast cancer in the first and third cycles of outpatient chemotherapy and to identify factors associated with it. METHODS The prospective longitudinal study was conducted at a Reference Hospital for Cancer Care in Brazil and included women aged ≥18 y diagnosed with stage I to III breast cancer receiving outpatient chemotherapy. Assessments were performed during the 1st and 3rd cycles of chemotherapy, including anthropometric measurements, sociodemographic data, clinical information, and quality of life. Nutritional risk was assessed using the NRS-2002. RESULTS Overweight was predominant in both chemotherapy cycles. Approximately 6.67% and 10% of patients were at nutritional risk in the 1st and 3rd chemotherapy cycles, respectively. Anxiety/depression was prevalent in the 1st chemotherapy cycle and was significantly associated with nutritional risk (P = 0.002). The variables age in cycle 3 and pain/discomfort in cycle 1 (P = 0.049 and P = 0.043, respectively) showed a significant association with nutritional risk. CONCLUSIONS This study highlights the complex interaction between nutritional status, neuropsychological symptoms, and sociodemographic characteristics in breast cancer patients during chemotherapy, and underscores the need for personalized interventions to improve oncological care.
Collapse
Affiliation(s)
| | - Wesley Rocha Grippa
- Graduate Program in Public Health, Federal University of Espírito Santo (UFES), Vitoria, ES, Brazil
| | | | | | - Andressa Bolsoni-Lopes
- Graduate Program in Nutrition and Health, Federal University of Espírito Santo (UFES), Vitoria, ES, Brazil
| | - Karolini Zuqui Nunes
- Graduate Program in Nutrition and Health, Federal University of Espírito Santo (UFES), Vitoria, ES, Brazil; Graduate Program in Public Health, Federal University of Espírito Santo (UFES), Vitoria, ES, Brazil
| | - Luís Carlos Lopes-Júnior
- Graduate Program in Nutrition and Health, Federal University of Espírito Santo (UFES), Vitoria, ES, Brazil; Graduate Program in Public Health, Federal University of Espírito Santo (UFES), Vitoria, ES, Brazil.
| |
Collapse
|
25
|
Anazco D, Acosta A, Cathcart-Rake EJ, D'Andre SD, Hurtado MD. Weight-centric prevention of cancer. OBESITY PILLARS 2024; 10:100106. [PMID: 38495815 PMCID: PMC10943063 DOI: 10.1016/j.obpill.2024.100106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 02/29/2024] [Accepted: 02/29/2024] [Indexed: 03/19/2024]
Abstract
Background The link between excess adiposity and carcinogenesis has been well established for multiple malignancies, and cancer is one of the main contributors to obesity-related mortality. The potential role of different weight-loss interventions on cancer risk modification has been assessed, however, its clinical implications remain to be determined. In this clinical review, we present the data assessing the effect of weight loss interventions on cancer risk. Methods In this clinical review, we conducted a comprehensive search of relevant literature using MEDLINE, Embase, Web of Science, and Google Scholar databases for relevant studies from inception to January 20, 2024. In this clinical review, we present systematic reviews and meta-analysis, randomized clinical trials, and prospective and retrospective observational studies that address the effect of different treatment modalities for obesity in cancer risk. In addition, we incorporate the opinions from experts in the field of obesity medicine and oncology regarding the potential of weight loss as a preventative intervention for cancer. Results Intentional weight loss achieved through different modalities has been associated with a reduced cancer incidence. To date, the effect of weight loss on the postmenopausal women population has been more widely studied, with multiple reports indicating a protective effect of weight loss on hormone-dependent malignancies. The effect of bariatric interventions as a protective intervention for cancer has been studied extensively, showing a significant reduction in cancer incidence and mortality, however, data for the effect of bariatric surgery on certain specific types of cancer is conflicting or limited. Conclusion Medical nutrition therapy, exercise, antiobesity medication, and bariatric interventions, might lead to a reduction in cancer risk through weight loss-dependent and independent factors. Further evidence is needed to better determine which population might benefit the most, and the amount of weight loss required to provide a clinically significant preventative effect.
Collapse
Affiliation(s)
- Diego Anazco
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Andres Acosta
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | | | | | - Maria D. Hurtado
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Department of Medicine, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
26
|
Chandrasekaran P, Weiskirchen R. The signaling pathways in obesity-related complications. J Cell Commun Signal 2024; 18:e12039. [PMID: 38946722 PMCID: PMC11208128 DOI: 10.1002/ccs3.12039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 07/02/2024] Open
Abstract
Obesity, a rapidly expanding epidemic worldwide, is known to exacerbate many medical conditions, making it a significant factor in multiple diseases and their associated complications. This threatening epidemic is linked to various harmful conditions such as type 2 diabetes mellitus, hypertension, metabolic dysfunction-associated steatotic liver disease, polycystic ovary syndrome, cardiovascular diseases (CVDs), dyslipidemia, and cancer. The rise in urbanization and sedentary lifestyles creates an environment that fosters obesity, leading to both psychosocial and medical complications. To identify individuals at risk and ensure timely treatment, it is crucial to have a better understanding of the pathophysiology of obesity and its comorbidities. This comprehensive review highlights the relationship between obesity and obesity-associated complications, including type 2 diabetes, hypertension, (CVDs), dyslipidemia, polycystic ovary syndrome, metabolic dysfunction-associated steatotic liver disease, gastrointestinal complications, and obstructive sleep apnea. It also explores the potential mechanisms underlying these associations. A thorough analysis of the interplay between obesity and its associated complications is vital in developing effective therapeutic strategies to combat the exponential increase in global obesity rates and mitigate the deadly consequences of this polygenic condition.
Collapse
Affiliation(s)
| | - Ralf Weiskirchen
- Institute of Molecular PathobiochemistryExperimental Gene Therapy and Clinical Chemistry (IFMPEGKC)RWTH University Hospital AachenAachenGermany
| |
Collapse
|
27
|
Albers FEM, Lou MWC, Dashti SG, Swain CTV, Rinaldi S, Viallon V, Karahalios A, Brown KA, Gunter MJ, Milne RL, English DR, Lynch BM. Sex-steroid hormones and risk of postmenopausal estrogen receptor-positive breast cancer: a case-cohort analysis. Cancer Causes Control 2024; 35:921-933. [PMID: 38363402 PMCID: PMC11130059 DOI: 10.1007/s10552-024-01856-6] [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: 01/16/2024] [Indexed: 02/17/2024]
Abstract
PURPOSE Sex-steroid hormones are associated with postmenopausal breast cancer but potential confounding from other biological pathways is rarely considered. We estimated risk ratios for sex-steroid hormone biomarkers in relation to postmenopausal estrogen receptor (ER)-positive breast cancer, while accounting for biomarkers from insulin/insulin-like growth factor-signaling and inflammatory pathways. METHODS This analysis included 1208 women from a case-cohort study of postmenopausal breast cancer within the Melbourne Collaborative Cohort Study. Weighted Poisson regression with a robust variance estimator was used to estimate risk ratios (RRs) and 95% confidence intervals (CIs) of postmenopausal ER-positive breast cancer, per doubling plasma concentration of progesterone, estrogens, androgens, and sex-hormone binding globulin (SHBG). Analyses included sociodemographic and lifestyle confounders, and other biomarkers identified as potential confounders. RESULTS Increased risks of postmenopausal ER-positive breast cancer were observed per doubling plasma concentration of progesterone (RR: 1.22, 95% CI 1.03 to 1.44), androstenedione (RR 1.20, 95% CI 0.99 to 1.45), dehydroepiandrosterone (RR: 1.15, 95% CI 1.00 to 1.34), total testosterone (RR: 1.11, 95% CI 0.96 to 1.29), free testosterone (RR: 1.12, 95% CI 0.98 to 1.28), estrone (RR 1.21, 95% CI 0.99 to 1.48), total estradiol (RR 1.19, 95% CI 1.02 to 1.39) and free estradiol (RR 1.22, 95% CI 1.05 to 1.41). A possible decreased risk was observed for SHBG (RR 0.83, 95% CI 0.66 to 1.05). CONCLUSION Progesterone, estrogens and androgens likely increase postmenopausal ER-positive breast cancer risk, whereas SHBG may decrease risk. These findings strengthen the causal evidence surrounding the sex-hormone-driven nature of postmenopausal breast cancer.
Collapse
Affiliation(s)
- Frances E M Albers
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Australia
- Cancer Epidemiology Division, Cancer Council Victoria, Council Victoria, Level 8, 200 Victoria Parade, East Melbourne, Melbourne, VIC, 3002, Australia
| | - Makayla W C Lou
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Australia
- Cancer Epidemiology Division, Cancer Council Victoria, Council Victoria, Level 8, 200 Victoria Parade, East Melbourne, Melbourne, VIC, 3002, Australia
| | - S Ghazaleh Dashti
- Clinical Epidemiology and Biostatistics Unit, Murdoch Children's Research Institute, Melbourne, Australia
- Department of Paediatrics, Melbourne Medical School, University of Melbourne, Melbourne, Australia
| | - Christopher T V Swain
- Cancer Epidemiology Division, Cancer Council Victoria, Council Victoria, Level 8, 200 Victoria Parade, East Melbourne, Melbourne, VIC, 3002, Australia
- Department of Physiotherapy, Melbourne School of Health Sciences, University of Melbourne, Melbourne, Australia
| | - Sabina Rinaldi
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, Lyon, France
| | - Vivian Viallon
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, Lyon, France
| | - Amalia Karahalios
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Australia
| | - Kristy A Brown
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, USA
| | - Marc J Gunter
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, Lyon, France
- Cancer Epidemiology and Prevention Research Unit, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Roger L Milne
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Australia
- Cancer Epidemiology Division, Cancer Council Victoria, Council Victoria, Level 8, 200 Victoria Parade, East Melbourne, Melbourne, VIC, 3002, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Melbourne, Australia
| | - Dallas R English
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Australia
- Cancer Epidemiology Division, Cancer Council Victoria, Council Victoria, Level 8, 200 Victoria Parade, East Melbourne, Melbourne, VIC, 3002, Australia
| | - Brigid M Lynch
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Australia.
- Cancer Epidemiology Division, Cancer Council Victoria, Council Victoria, Level 8, 200 Victoria Parade, East Melbourne, Melbourne, VIC, 3002, Australia.
- Physical Activity Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia.
| |
Collapse
|
28
|
Han DS, Lee EO. Leptin Promotes Vasculogenic Mimicry in Breast Cancer Cells by Regulating Aquaporin-1. Int J Mol Sci 2024; 25:5215. [PMID: 38791252 PMCID: PMC11121373 DOI: 10.3390/ijms25105215] [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: 04/25/2024] [Revised: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
Leptin is an obesity-related hormone that plays an important role in breast cancer progression. Vasculogenic mimicry (VM) refers to the formation of vascular channels lined by tumor cells. This study aimed to investigate the relationship between leptin and VM in human breast cancer cells. VM was measured by a 3D culture assay. Signal transducers and activators of transcription 3 (STAT3) signaling, aquaporin-1 (AQP1), and the expression of VM-related proteins, including vascular endothelial cadherin (VE-cadherin), twist, matrix metalloproteinase-2 (MMP-2), and laminin subunit 5 gamma-2 (LAMC2), were examined by Western blot. AQP1 mRNA was analyzed by a reverse transcriptase-polymerase chain reaction (RT-PCR). Leptin increased VM and upregulated phospho-STAT3, VE-cadherin, twist, MMP-2, and LAMC2. These effects were inhibited by the leptin receptor-blocking peptide, Ob-R BP, and the STAT3 inhibitor, AG490. A positive correlation between leptin and AQP1 mRNA was observed and was confirmed by RT-PCR. Leptin upregulated AQP1 expression, which was blocked by Ob-R BP and AG490. AQP1 overexpression increased VM and the expression of VM-related proteins. AQP1 silencing inhibited leptin-induced VM and the expression of VM-related proteins. Thus, these results showed that leptin facilitates VM in breast cancer cells via the Ob-R/STAT3 pathway and that AQP1 is a key mediator in leptin-induced VM.
Collapse
Affiliation(s)
- Deok-Soo Han
- Department of Science in Korean Medicine, College of Korean Medicine, Graduate School, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea;
| | - Eun-Ok Lee
- Department of Science in Korean Medicine, College of Korean Medicine, Graduate School, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea;
- Department of Cancer Preventive Material Development, College of Korean Medicine, Graduate School, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| |
Collapse
|
29
|
SenthilKumar G, Schottstaedt AM, Peterson LL, Pedersen LN, Chitambar CR, Vistocky A, Banerjee A, Longo JM, Kelly T, Currey A, Stolley MR, Bergom C. Stay on Track: A Pilot Randomized Control Trial on the Feasibility of a Diet and Exercise Intervention in Patients with Breast Cancer Receiving Radiotherapy. CANCER RESEARCH COMMUNICATIONS 2024; 4:1211-1226. [PMID: 38530195 PMCID: PMC11075661 DOI: 10.1158/2767-9764.crc-23-0148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/14/2023] [Accepted: 03/18/2024] [Indexed: 03/27/2024]
Abstract
PURPOSE Among patients with breast cancer undergoing radiotherapy, posttreatment cardiovascular disease and worsened quality of life (QoL) are leading causes of morbidity and mortality. To overcome these negative radiotherapy effects, this prospective, randomized clinical trial pilots a 12-week Stay on Track exercise and diet intervention for overweight patients with nonmetastatic breast cancer undergoing whole-breast radiotherapy. EXPERIMENTAL DESIGN The intervention group (n = 22) participated in three personal exercise and dietary counseling sessions, and received three text reminders/week to adhere to recommendations. The control group (n = 22) was administered a diet/exercise information binder. All patients received a Fitbit, and at baseline, 3 months, and 6 months, measurements of biomarkers, dual-energy X-ray absorptiometry scans, QoL and physical activity surveys, and food frequency questionnaires were obtained. A satisfaction survey was administered at 3 months. RESULTS Stay on Track was well received, with high rates of adherence and satisfaction. The intervention group showed an increase in self-reported physical activity and preserved QoL, a decrease in body mass index and visceral fat, and higher American Cancer Society/American Institute of Cancer Research dietary adherence. The control participants had reduced QoL, anti-inflammatory markers, and increased metabolic syndrome markers. Both groups had decreased overall body mass. These changes were within group effects. When comparing the intervention and control groups over time, there were notable improvements in dietary adherence in the intervention group. CONCLUSIONS Targeted lifestyle interventions during radiotherapy are feasible and could decrease cardiovascular comorbidities in patients with breast cancer. Larger-scale implementation with longer follow-up can better determine interventions that influence cardiometabolic health and QoL. SIGNIFICANCE This pilot study examines cardiometabolic benefits of a combined diet and exercise intervention for patients with breast cancer undergoing radiotherapy. The intervention included an activity tracker (FitBit) and text message reminders to promote adherence to lifestyle interventions. Large-scale implementation of such programs may improve cardiometabolic outcomes and overall QoL among patients with breast cancer.
Collapse
Affiliation(s)
- Gopika SenthilKumar
- Department of Physiology and Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | | | - Lindsay L. Peterson
- Division of Medical Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
- Alvin J. Siteman Cancer Center, Washington University in St. Louis, St. Louis, Missouri
| | - Lauren N. Pedersen
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Christopher R. Chitambar
- Division of Medical Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Alexis Vistocky
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Anjishnu Banerjee
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - John M. Longo
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Tracy Kelly
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Adam Currey
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Melinda R. Stolley
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Carmen Bergom
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
- Alvin J. Siteman Cancer Center, Washington University in St. Louis, St. Louis, Missouri
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
30
|
Abd El-Hack ME, Kamal M, Alazragi RS, Alreemi RM, Qadhi A, Ghafouri K, Azhar W, Shakoori AM, Alsaffar N, Naffadi HM, Taha AE, Abdelnour SA. Impacts of chitosan and its nanoformulations on the metabolic syndromes: a review. BRAZ J BIOL 2024; 83:e276530. [PMID: 38422267 DOI: 10.1590/1519-6984.276530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 11/15/2023] [Indexed: 03/02/2024] Open
Abstract
A significant public health issue worldwide is metabolic syndrome, a cluster of metabolic illnesses that comprises insulin resistance, obesity, dyslipidemia, hyperglycemia, and hypertension. The creation of natural treatments and preventions for metabolic syndrome is crucial. Chitosan, along with its nanoformulations, is an oligomer of chitin, the second-most prevalent polymer in nature, which is created via deacetylation. Due to its plentiful biological actions in recent years, chitosan and its nanoformulations have drawn much interest. Recently, the chitosan nanoparticle-based delivery of CRISPR-Cas9 has been applied in treating metabolic syndromes. The benefits of chitosan and its nanoformulations on insulin resistance, obesity, diabetes mellitus, dyslipidemia, hyperglycemia, and hypertension will be outlined in the present review, highlighting potential mechanisms for the avoidance and medication of the metabolic syndromes by chitosan and its nanoformulations.
Collapse
Affiliation(s)
- M E Abd El-Hack
- Zagazig University, Faculty of Agriculture, Department of Poultry, Zagazig, Egypt
| | - M Kamal
- Agricultural Research Center, Animal Production Research Institute, Dokki, Giza, Egypt
| | - R S Alazragi
- University of Jeddah, College of Science, Department of Biochemistry, Jeddah, Saudi Arabia
| | - R M Alreemi
- University of Jeddah, College of Science, Department of Biochemistry, Jeddah, Saudi Arabia
| | - A Qadhi
- Umm Al-Qura University, Faculty of Applied Medical Sciences, Clinical Nutrition Department, Makkah, Saudi Arabia
| | - K Ghafouri
- Umm Al-Qura University, Faculty of Applied Medical Sciences, Clinical Nutrition Department, Makkah, Saudi Arabia
| | - W Azhar
- Umm Al-Qura University, Faculty of Applied Medical Sciences, Clinical Nutrition Department, Makkah, Saudi Arabia
| | - A M Shakoori
- Umm Al-Qura University, Faculty of Applied Medical Sciences, Laboratory Medicine Department, Makkah, Kingdom of Saudi Arabia
| | - N Alsaffar
- Mohammed Al-Mana College for Medical Sciences, Biochemistry and Molecular Biology Department, Dammam, Saudi Arabia
| | - H M Naffadi
- Umm Al-Qura University, College of Medicine, Department of Medical Genetics, Makkah, Kingdom of Saudi Arabia
| | - A E Taha
- Alexandria University, Faculty of Veterinary Medicine, Department of Animal Husbandry and Animal Wealth Development, Edfina, Egypt
| | - S A Abdelnour
- Zagazig University, Faculty of Agriculture, Department of Animal Production, Zagazig, Egypt
| |
Collapse
|
31
|
Song Q, Muller KE, Hondelink LM, diFlorio-Alexander RM, Karagas MR, Hassanpour S. Nonmetastatic Axillary Lymph Nodes Have Distinct Morphology and Immunophenotype in Obese Patients with Breast Cancer at Risk for Metastasis. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:253-263. [PMID: 38029922 PMCID: PMC10835463 DOI: 10.1016/j.ajpath.2023.11.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 11/02/2023] [Accepted: 11/07/2023] [Indexed: 12/01/2023]
Abstract
Obese patients with breast cancer have worse outcomes than their normal weight counterparts, with a 50% to 80% increased rate of axillary nodal metastasis. Recent studies suggest a link between increased lymph node adipose tissue and breast cancer nodal metastasis. Further investigation into potential mechanisms underlying this link may reveal potential prognostic utility of fat-enlarged lymph nodes in patients with breast cancer. This study used a deep learning model to identify morphologic differences in nonmetastatic axillary nodes between obese, node-positive, and node-negative patients with breast cancer. The model was developed using nested cross-validation on 180 cases and achieved an area under the receiver operator characteristic curve of 0.67 in differentiating patients using hematoxylin and eosin-stained whole slide images. The morphologic analysis of the predictive regions showed an increased average adipocyte size (P = 0.004), increased white space between lymphocytes (P < 0.0001), and increased red blood cells (P < 0.001) in nonmetastatic lymph nodes of node-positive patients. Preliminary immunohistochemistry analysis on a subset of 30 patients showed a trend of decreased CD3 expression and increased leptin expression in fat-replaced axillary lymph nodes of obese, node-positive patients. These findings suggest a novel direction to further investigate the interaction between lymph node adiposity, lymphatic dysfunction, and breast cancer nodal metastases, highlighting a possible prognostic tool for obese patients with breast cancer.
Collapse
Affiliation(s)
- Qingyuan Song
- Department of Biomedical Data Science, Dartmouth College, Hanover, New Hampshire
| | - Kristen E Muller
- Department of Pathology and Laboratory Medicine, Dartmouth Health, Lebanon, New Hampshire
| | - Liesbeth M Hondelink
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | | | | | - Saeed Hassanpour
- Department of Biomedical Data Science, Dartmouth College, Hanover, New Hampshire; Department of Epidemiology, Dartmouth College, Hanover, New Hampshire; Department of Computer Science, Dartmouth College, Hanover, New Hampshire.
| |
Collapse
|
32
|
Ray I, Möller-Levet CS, Michael A, Butler-Manuel S, Chatterjee J, Tailor A, Ellis PE, Meira LB. Circulating Adipocytokines and Insulin Like-Growth Factors and Their Modulation in Obesity-Associated Endometrial Cancer. Cancers (Basel) 2024; 16:531. [PMID: 38339282 PMCID: PMC10854745 DOI: 10.3390/cancers16030531] [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: 11/30/2023] [Revised: 01/13/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
The rising global incidence of uterine cancer is linked to the escalating prevalence of obesity. Obesity results in alterations in adipocytokines and IGFs, driving cancer progression via inflammation, increased cell proliferation, and apoptosis inhibition, although the precise mechanisms are still unclear. This study examined a set of six markers, namely, adiponectin, leptin, IL6, TNFα, IGF1, and IGF2 and compared them between fifty age-matched endometrial cancer patients (study group) and non-cancer patients with benign gynaecological conditions (control group). We also assessed the relationship of these markers with obesity and explored the correlation between these markers and various tumour characteristics. In the cancer population, these markers were also assessed 24 h and 6 months post-surgery. Remarkably, low adiponectin levels were associated with a 35.8% increase in endometrial cancer risk. Interestingly, compared to control subjects where IGF levels decreased after menopause, post-menopausal women in the study group showed elevated IGF1 and IGF2 levels, suggesting a potential influence of endometrial cancer on the IGF system, particularly after menopause. Lastly, it is noteworthy that a discernible inverse relationship trend was observed in the levels of adipocytokines and IGFs 6 months post-surgery. This indicates that treatment for endometrial cancer may have a differential impact on adipocytokines and IGFs, potentially holding clinical significance that merits further investigation.
Collapse
Affiliation(s)
- Irene Ray
- Department of Clinical and Experimental Medicine, University of Surrey, Daphne Jackson Road, Guildford GU2 7WG, UK
- Academic Department of Gynaecological Oncology, Royal Surrey NHS Foundation Trust, Egerton Road, Guildford GU2 7XX, UK
| | - Carla S. Möller-Levet
- Bioinformatics Core Facility, University of Surrey, Daphne Jackson Road, Guildford GU2 7WG, UK
| | - Agnieszka Michael
- Department of Clinical and Experimental Medicine, University of Surrey, Daphne Jackson Road, Guildford GU2 7WG, UK
- Department of Oncology, Royal Surrey NHS Foundation Trust, Egerton Road, Guildford GU2 7XX, UK
| | - Simon Butler-Manuel
- Academic Department of Gynaecological Oncology, Royal Surrey NHS Foundation Trust, Egerton Road, Guildford GU2 7XX, UK
| | - Jayanta Chatterjee
- Academic Department of Gynaecological Oncology, Royal Surrey NHS Foundation Trust, Egerton Road, Guildford GU2 7XX, UK
- Department of Life Sciences, Brunel University London, Kingston Lane Uxbridge, Middlesex, Uxbridge UB8 3PH, UK
| | - Anil Tailor
- Academic Department of Gynaecological Oncology, Royal Surrey NHS Foundation Trust, Egerton Road, Guildford GU2 7XX, UK
| | - Patricia E. Ellis
- Department of Clinical and Experimental Medicine, University of Surrey, Daphne Jackson Road, Guildford GU2 7WG, UK
- Academic Department of Gynaecological Oncology, Royal Surrey NHS Foundation Trust, Egerton Road, Guildford GU2 7XX, UK
| | - Lisiane B. Meira
- Department of Clinical and Experimental Medicine, University of Surrey, Daphne Jackson Road, Guildford GU2 7WG, UK
| |
Collapse
|
33
|
Bou Malhab LJ, Nair VA, Qaisar R, Pintus G, Abdel-Rahman WM. Towards Understanding the Development of Breast Cancer: The Role of RhoJ in the Obesity Microenvironment. Cells 2024; 13:174. [PMID: 38247865 PMCID: PMC10814036 DOI: 10.3390/cells13020174] [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: 10/25/2023] [Revised: 12/21/2023] [Accepted: 01/04/2024] [Indexed: 01/23/2024] Open
Abstract
Obesity is a growing pandemic with an increasing risk of inducing different cancer types, including breast cancer. Adipose tissue is proposed to be a major player in the initiation and progression of breast cancer in obese people. However, the mechanistic link between adipogenicity and tumorigenicity in breast tissues is poorly understood. We used in vitro and in vivo approaches to investigate the mechanistic relationship between obesity and the onset and progression of breast cancer. In obesity, adipose tissue expansion and remodeling are associated with increased inflammatory mediator's release and anti-inflammatory mediators' reduction.. In order to mimic the obesity micro-environment, we cultured cells in an enriched pro-inflammatory cytokine medium to which we added a low concentration of beneficial adipokines. Epithelial cells exposed to the obesity micro-environment were phenotypically transformed into mesenchymal-like cells, characterized by an increase in different mesenchymal markers and the acquisition of the major hallmarks of cancerous cells; these include sustained DNA damage, the activation of the ATR-Chk2 pathway, an increase in proliferation rate, cell invasion, and resistance to conventional chemotherapy. Transcriptomic analysis revealed that several genes, including RhoJ, CCL7, and MMP9, acted as potential major players in the observed phenomenon. The transcriptomics findings were confirmed in vitro using qRT-PCR and in vivo using high-fat-diet-fed mice. Our data suggests RhoJ as a potential novel molecular driver of tumor development in breast tissues and a mediator of cell resistance to conventional chemotherapy through PAK1 activation. These data propose that RhoJ is a potential target for therapeutic interventions in obese breast cancer patients.
Collapse
Affiliation(s)
- Lara J. Bou Malhab
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates;
| | - Vidhya A. Nair
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates;
| | - Rizwan Qaisar
- Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates;
| | - Gianfranco Pintus
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy;
| | - Wael M. Abdel-Rahman
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates;
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| |
Collapse
|
34
|
Šenigl F, Soikkeli A, Prost S, Schatz DG, Slavková M, Hejnar J, Alinikula J. The SV40 virus enhancer functions as a somatic hypermutation-targeting element with potential oncogenic activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.09.574829. [PMID: 38260396 PMCID: PMC10802419 DOI: 10.1101/2024.01.09.574829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Simian virus 40 (SV40) is a monkey virus associated with several types of human cancers. SV40 is most frequently detected in mesotheliomas, brain and bone tumors and lymphomas, but the mechanism for SV40 tumorigenesis in humans is not clear. SV40 relative Merkel cell polyomavirus (MCPyV) causes Merkel cell carcinoma (MCC) in humans by expressing truncated large tumor antigen (LT) caused by APOBEC cytidine deaminase family enzymes induced mutations. AID (activation-induced cytidine deaminase), a member of the APOBEC family, is the initiator of the antibody diversification process known as somatic hypermutation (SHM) and its aberrant expression and targeting is a frequent source of lymphomagenesis. In this study, we investigated whether AID-induced mutations could cause truncation of SV40 LT. We demonstrate that the SV40 enhancer has strong SHM targeting activity in several cell types and that AID-induced mutations accumulate to SV40 LT in B cells and kidney cells and cause truncated LT expression in B cells. Our results argue that the ability of the SV40 enhancer to target SHM to LT is a potential source of LT truncation events in various cell types that could contribute to carcinogenesis.
Collapse
|
35
|
Kim JW, Kim JH, Lee YJ. The Role of Adipokines in Tumor Progression and Its Association with Obesity. Biomedicines 2024; 12:97. [PMID: 38255203 PMCID: PMC10813163 DOI: 10.3390/biomedicines12010097] [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: 10/29/2023] [Revised: 12/26/2023] [Accepted: 12/27/2023] [Indexed: 01/24/2024] Open
Abstract
Obesity is a well-established risk factor for various malignancies and emerging evidence suggests that adipokines play a pivotal role in linking excess adiposity to tumorigenesis. Adipokines are bioactive molecules secreted by adipose tissue and their altered expression in obesity contributes to a pro-inflammatory, pro-angiogenic, and growth-promoting microenvironment conducive to tumorigenesis. Leptin, a key adipokine, activates survival and proliferative signaling pathways whereas adiponectin exhibits tumor-suppressive effects by inducing apoptosis and cell cycle arrest. Visfatin has also been documented to promote tumor growth, angiogenesis, migration, and invasion. Moreover, emerging studies suggest that adipokines, such as resistin, apelin, and chemerin, which are overexpressed in obesity, may also possess oncogenic functions. Despite advancements in our understanding of the roles of individual adipokines in cancer, the intricate interplay and crosstalk between adipokines, tumor cells, and the tumor microenvironment remain complex and multifaceted. This review highlights the evolving knowledge of how adipokines contribute to obesity-related tumorigenesis, shedding light on the potential of targeting adipokine signaling pathways as a novel therapeutic approach for obesity-associated cancers. Further research on the specific mechanisms and interactions between adipokines and tumor cells is crucial for a comprehensive understanding of obesity-associated cancer pathogenesis.
Collapse
Affiliation(s)
| | | | - Yoon Jae Lee
- Department of Plastic and Reconstructive Surgery, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 07345, Republic of Korea; (J.W.K.); (J.H.K.)
| |
Collapse
|
36
|
Neagu M, Dobre EG. New Insights into the Link Between Melanoma and Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:851-867. [PMID: 39287874 DOI: 10.1007/978-3-031-63657-8_28] [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: 09/19/2024]
Abstract
The significant increase in the incidence of obesity represents a global health crisis. Obesity is actually a multi-organ disease affecting the entire organism; hence, skin is no exception. As the functional alterations in the adipose tissue are contributing factors to many diseases, including cancer, recently, the link between the development of melanoma skin cancer and obesity gains increased attention. Besides several other factors, the increase of adipose stromal/stem cells (ASCs) impacts cancer progression. Moreover, increased production of cytokines and growth factors done by ASCs induces tumorigenesis and metastasis. The chronic inflammatory state that is sustained by this metabolic imbalance favors skin malignancies, melanoma included. Cutaneous melanoma, as an aggressive skin cancer, has both intrinsic and extrinsic risk factors where sun exposure and lifestyles are the main environmental factors inducing this skin cancer. With the advent of recent targeted and immune-based therapies in melanoma, the link between obesity and the efficacy of these therapies in melanoma remains controversial. A recent molecular relationship between the melanocortin pathway appending to both melanin synthesis and obesity was established. The biology of adipokines, molecules secreted by the adipose tissue, is linked to inflammation, and their molecular pathways can be involved in angiogenesis, migration, invasion, and proliferation of melanoma cells. In melanoma cells, among the most noticeable metabolic reprogramming characteristics is an increased rate of lipid synthesis. Lipid mediators impact classical oncogenic pathways, affecting melanoma progression. The chapter will tackle also the practical implications for melanoma prevention and treatment, namely, how metabolic manipulation can be exploited to overcome immunosuppression and support immune checkpoint blockade efficacy.
Collapse
Affiliation(s)
- Monica Neagu
- Immunology Laboratory, Victor Babes National Institute of Pathology, Bucharest, Romania
- Pathology Department, Colentina University Hospital, Bucharest, Romania
- Doctoral School of Biology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Elena-Georgiana Dobre
- Immunology Laboratory, Victor Babes National Institute of Pathology, Bucharest, Romania
| |
Collapse
|
37
|
Lee CM, Fang S. Fat Biology in Triple-Negative Breast Cancer: Immune Regulation, Fibrosis, and Senescence. J Obes Metab Syndr 2023; 32:312-321. [PMID: 38014425 PMCID: PMC10786212 DOI: 10.7570/jomes23044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/18/2023] [Accepted: 11/21/2023] [Indexed: 11/29/2023] Open
Abstract
Obesity, now officially recognized as a disease requiring intervention, has emerged as a significant health concern due to its strong association with elevated susceptibility to diverse diseases and various types of cancer, including breast cancer. The link between obesity and cancer is intricate, with obesity exerting a significant impact on cancer recurrence and elevated mortality rates. Among the various subtypes of breast cancer, triple-negative breast cancer (TNBC) is the most aggressive, accounting for 15% to 20% of all cases. TNBC is characterized by low expression of estrogen receptors and progesterone receptors as well as the human epidermal growth factor 2 receptor protein. This subtype poses distinct challenges in terms of treatment response and exhibits strong invasiveness. Furthermore, TNBC has garnered attention because of its association with obesity, in which excess body fat and reduced physical activity have been identified as contributing factors to the increased incidence of this aggressive form of breast cancer. In this comprehensive review, the impact of obesity on TNBC was explored. Specifically, we focused on the three key mechanisms by which obesity affects TNBC development and progression: modification of the immune profile, facilitation of fibrosis, and initiation of senescence. By comprehensively examining these mechanisms, we illuminated the complex interplay between TNBC and obesity, facilitating the development of novel approaches for prevention, early detection, and effective management of this challenging disease.
Collapse
Affiliation(s)
- Chae Min Lee
- Graduate School of Medical Science, Brain Korea 2 Project, Yonsei University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Sungsoon Fang
- Graduate School of Medical Science, Brain Korea 2 Project, Yonsei University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, Korea
- Severance Institute for Vascular and Metabolic Research, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
38
|
赵 欢, 凌 羽, 宓 帅, 朱 家, 范 佳, 杨 叶, 王 晶, 李 迎. [Associations of circulating leptin levels with colorectal adenoma and colorectal cancer: a case-control and Mendelian randomization study]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2023; 43:1989-1997. [PMID: 38189383 PMCID: PMC10774100 DOI: 10.12122/j.issn.1673-4254.2023.12.01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Indexed: 01/09/2024]
Abstract
OBJECTIVE To explore the causal association between circulating leptin levels and the risk of colorectal adenoma and colorectal cancer. METHODS We collected demographic and clinical data and serum samples from 497 patients with colorectal adenoma, 955 patients with colorectal cancer, and 911 healthy individuals from the First Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang Cancer Hospital, Zhuji People's Hospital, and Lin'an District First People's Hospital. Instrumental variables of leptin were selected and genotyping tests were performed. A logistic regression model and stratified analysis were used to evaluate the association of serum leptin levels with colorectal adenoma, colorectal cancer, and the progression of colorectal adenoma to colorectal cancer. Genetic risk score (GRS) and single nucleotide polymorphisms (SNPs) were further used as instrumental variables in one-sample and two-sample Mendelian randomization analyses leveraging two-stage least squares and inverse-variance weighted methods to estimate the causal association of leptin levels with the risk of colorectal adenoma, colorectal cancer, and progression of colorectal adenoma to colorectal cancer. RESULTS High levels of leptin, compared with its lowest quartile, were positively correlated with colorectal adenoma (P=0.005) and negatively with colorectal cancer (P < 0.001) and the risk of progression of colorectal adenoma to colorectal cancer (P < 0.001). Mendelian randomization analysis showed that GRS of leptin, either weighted or not, was not significantly correlated with the risk of colorectal adenoma, colorectal cancer, or the progression of colorectal adenoma to colorectal cancer, nor did the two-sample Mendelian randomization study support an association between leptin and the risk of colorectal cancer (P>0.05). CONCLUSION Although the case-control study suggests probable correlations of leptin with the risk of colorectal adenoma, colorectal cancer, and colorectal adenoma progression to colorectal cancer, Mendelian randomization studies did not support a causal association of leptin with the risks of colorectal adenoma, colorectal cancer, or colorectal adenoma progression to colorectal cancer.
Collapse
Affiliation(s)
- 欢灵 赵
- 杭州医学院公共卫生系流行病与卫生统计学教研室,浙江 杭州 310053Department of Epidemiology and Health Statistics, School of Public Health, Hangzhou Medical College, Hangzhou 310053, China
| | - 羽晓 凌
- 宁波大学医学院公共卫生学院,浙江 宁波 315211School of Public Health, School of Medicine, Ningbo University, Ningbo 315211, China
| | - 帅 宓
- 德清县莫干山镇卫生院,浙江 德清 313200Deqing County Moganshan Town Health Center, Deqing 313200, China
| | - 家豪 朱
- 杭州医学院公共卫生系流行病与卫生统计学教研室,浙江 杭州 310053Department of Epidemiology and Health Statistics, School of Public Health, Hangzhou Medical College, Hangzhou 310053, China
| | - 佳耀 范
- 浙江大学医学院公共卫生学院大数据健康科学系,浙江 杭州 310058Department of Big Data in Health Science, School of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - 叶 杨
- 杭州医学院公共卫生系流行病与卫生统计学教研室,浙江 杭州 310053Department of Epidemiology and Health Statistics, School of Public Health, Hangzhou Medical College, Hangzhou 310053, China
| | - 晶 王
- 杭州医学院公共卫生系流行病与卫生统计学教研室,浙江 杭州 310053Department of Epidemiology and Health Statistics, School of Public Health, Hangzhou Medical College, Hangzhou 310053, China
| | - 迎君 李
- 杭州医学院公共卫生系流行病与卫生统计学教研室,浙江 杭州 310053Department of Epidemiology and Health Statistics, School of Public Health, Hangzhou Medical College, Hangzhou 310053, China
| |
Collapse
|
39
|
Zhuang B, Zhang L, Wang Y, Cao Y, Shih Y, Jin S, Li H, Gong L, Wang Y, Jin S, Lu Q. Body composition and dietary intake in patients with head and neck cancer during radiotherapy: a longitudinal study. BMJ Support Palliat Care 2023; 13:445-452. [PMID: 32917650 DOI: 10.1136/bmjspcare-2020-002359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 06/14/2020] [Accepted: 08/11/2020] [Indexed: 12/24/2022]
Abstract
OBJECTIVES To investigate the body composition and dietary intake in the patients with head and neck cancer (HNC) during radiotherapy (RT), and explore the relationship between them. METHODS This was a prospective, longitudinal observational study. Adult patients with HNC undergoing RT between March 2017 and August 2018 were recruited. Patients' body compositions were evaluated by bioelectrical impedance analysis, and dietary intake was recorded by 24-hour dietary recall at three time points, including baseline (T1), mid-treatment (T2) and post-treatment (T3). Patients were divided into low, middle and high energy intake groups based on the average daily energy intake (DEI). Changes in body weight (BW), fat mass (FM), fat-free mass (FFM) and skeletal muscle mass (SMM) among these three groups were compared. RESULTS From T1 to T3, the median loss of patients' BW, FM, FFM and SMM was 4.60, 1.90, 2.60 and 1.50 kg, respectively. The loss of BW was more dramatic from T2 to T3 than that from T1 to T2. BW loss was mainly contributed by SMM loss from T1 to T2 and by FM loss from T2 to T3. Meanwhile, patients' dietary intake reduced during treatment. High DEI group had a significantly attenuated loss of patients' BW, FFM, SMM and FM compared with the low DEI group. CONCLUSION Patients' BW, FM, FFM and SMM all significantly reduced, especially from T2 to T3, with decreased DEI during RT, which stresses the importance of nutrition intervention during the whole course of RT.
Collapse
Affiliation(s)
- Bing Zhuang
- Division of Medical and Surgical Nursing, Peking University School of Nursing, Beijing, China
| | - Lichuan Zhang
- Division of Medical and Surgical Nursing, Peking University School of Nursing, Beijing, China
| | - Yujie Wang
- Division of Medical and Surgical Nursing, Peking University School of Nursing, Beijing, China
| | - Yiwei Cao
- Division of Medical and Surgical Nursing, Peking University School of Nursing, Beijing, China
| | - Yian Shih
- Division of Medical and Surgical Nursing, Peking University School of Nursing, Beijing, China
| | - Sanli Jin
- Division of Medical and Surgical Nursing, Peking University School of Nursing, Beijing, China
| | - Hongmei Li
- Division of Surgical Nursing, Shanxi Hospital of Integrated Traditional and Western Medicine, Taiyuan, China
| | - Liqing Gong
- Division of Clinical Nutrition, Beijing Cancer Hospital, Beijing, China
| | - Yanli Wang
- Division of Clinical Nutrition, Beijing Cancer Hospital, Beijing, China
| | - Shuai Jin
- Division of Medical and Surgical Nursing, Peking University School of Nursing, Beijing, China
| | - Qian Lu
- Division of Medical and Surgical Nursing, Peking University School of Nursing, Beijing, China
| |
Collapse
|
40
|
Silva RJG, Grippa WR, Neto LCBS, Enriquez-Martinez OG, Marcarini JAC, Pessanha RM, Haraguchi FK, Lopes-Júnior LC. Factors Associated with the Nutritional Status of Women with Non-Metastatic Breast Cancer in a Brazilian High Complexity Oncology Center. Nutrients 2023; 15:4961. [PMID: 38068818 PMCID: PMC10707825 DOI: 10.3390/nu15234961] [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: 07/29/2023] [Revised: 09/01/2023] [Accepted: 09/05/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Breast cancer poses a significant public health concern owing to its high prevalence and the risk of mortality associated with delayed diagnosis and treatment. The aim of this study was to assess the nutritional status of women with non-metastatic breast cancer and to identify factors associated with it. METHODS A cross-sectional observational study was conducted at a High Complexity Oncology Assistance Center in the southeast region of Brazil, with the aim of assessing the nutritional status in women undergoing treatment for stage I, II, or III breast cancer. Patients in palliative care or undergoing reconstructive surgery were excluded. Data collection took place between June 2022 and March 2023 and included questionnaires, physical examinations, laboratory tests, and anthropometric assessments. Nutritional status was assessed using measures such as BMI and skinfold thickness, while nutritional risk was assessed using the Nutritional Risk Screening (NRS-2002) tool. RESULTS Significant associations were found between nutritional risk and educational level (p = 0.03) and BMI (p = 0.01). Binary logistic regression analysis revealed a significant association between educational level and nutritional risk, indicating that lower educational level was associated with higher odds of nutritional risk (OR = 4.59; 95% CI = 1.01-21.04; p = 0.049). In addition, regarding BMI, it was observed that a BMI above 20.5 kg/m2 was associated with a higher likelihood of nutritional risk (OR = 0.09; 95% CI = 0.01-0.89; p = 0.039). CONCLUSIONS It is crucial to consider the nutritional status of breast cancer patients, alongside clinical factors, to offer comprehensive and personalized care. Gaining insight into the sociodemographic variables linked to nutritional risk can significantly contribute to our understanding of breast cancer. This knowledge, in turn, can aid in identifying effective strategies for public policy, health promotion, and prevention efforts aimed at tackling this condition.
Collapse
Affiliation(s)
- Roberto Júnio Gomes Silva
- Graduate Program in Nutrition and Health, Universidade Federal do Espírito Santo, Vitória 29500-000, ES, Brazil (F.K.H.)
| | - Wesley Rocha Grippa
- Graduate Program in Public Health, Universidade Federal do Espírito Santo, Vitória 29500-000, ES, Brazil; (W.R.G.); (R.M.P.)
| | | | | | | | - Raphael Manhães Pessanha
- Graduate Program in Public Health, Universidade Federal do Espírito Santo, Vitória 29500-000, ES, Brazil; (W.R.G.); (R.M.P.)
| | - Fabiano Kenji Haraguchi
- Graduate Program in Nutrition and Health, Universidade Federal do Espírito Santo, Vitória 29500-000, ES, Brazil (F.K.H.)
| | - Luís Carlos Lopes-Júnior
- Graduate Program in Nutrition and Health, Universidade Federal do Espírito Santo, Vitória 29500-000, ES, Brazil (F.K.H.)
- Graduate Program in Public Health, Universidade Federal do Espírito Santo, Vitória 29500-000, ES, Brazil; (W.R.G.); (R.M.P.)
| |
Collapse
|
41
|
Mubtasim N, Gollahon L. The Effect of Adipocyte-Secreted Factors in Activating Focal Adhesion Kinase-Mediated Cell Signaling Pathway towards Metastasis in Breast Cancer Cells. Int J Mol Sci 2023; 24:16605. [PMID: 38068928 PMCID: PMC10706115 DOI: 10.3390/ijms242316605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 11/07/2023] [Accepted: 11/17/2023] [Indexed: 12/18/2023] Open
Abstract
Obesity-associated perturbations in the normal secretion of adipocytokines from white adipocytes can drive the metastatic progression of cancer. However, the association between obesity-induced changes in secretory factors of white adipocytes and subsequent transactivation of the downstream effector proteins impacting metastasis in breast cancer cells remains unclear. Focal adhesion kinase, a cytoplasmic signal transducer, regulates the biological phenomenon of metastasis by activating downstream targets such as beta-catenin and MMP9. Thus, the possible role of phosphorylated FAK in potentiating cancer cell migration was investigated. To elucidate this potential relationship, MCF7 (ER+), MDA-MB-231 (Triple Negative) breast cancer cells, and MCF-10A non-tumorigenic breast cells were exposed to in vitro murine adipocyte-conditioned medium derived from 3T3-L1 MBX cells differentiated to obesity with fatty acid supplementation. Our results show that the conditioned medium derived from these obese adipocytes enhanced motility and invasiveness of breast cancer cells. Importantly, no such changes were observed in the non-tumorigenic breast cells. Our results also show that increased FAK autophosphorylation was followed by increased expression of beta-catenin and MMP9 in the breast cancer cells when exposed to obese adipocyte-conditioned medium, but not in the MCF10A cells. These results indicate that adipocyte-derived secretory factors induced FAK activation through phosphorylation. This in turn increased breast cancer cell migration and invasion by activating its downstream effector proteins beta-catenin and MMP9.
Collapse
Affiliation(s)
- Noshin Mubtasim
- Department of Biological Sciences, Texas Tech University, 2500 Broadway, Lubbock, TX 79409, USA;
| | - Lauren Gollahon
- Department of Biological Sciences, Texas Tech University, 2500 Broadway, Lubbock, TX 79409, USA;
- Obesity Research Institute, Texas Tech University, 2500 Broadway, Lubbock, TX 79409, USA
| |
Collapse
|
42
|
Sánchez-Jiménez F, Jiménez-Cortegana C. The obesity paradox. Med Clin (Barc) 2023; 161:342-343. [PMID: 37858344 DOI: 10.1016/j.medcli.2023.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 09/13/2023] [Indexed: 10/21/2023]
Affiliation(s)
- Flora Sánchez-Jiménez
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Seville, Spain; Clinical Biochemistry Service, Virgen Macarena University Hospital, Seville, Spain
| | - Carlos Jiménez-Cortegana
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Seville, Spain.
| |
Collapse
|
43
|
Albers FE, Lou MW, Dashti SG, Swain CT, Rinaldi S, Viallon V, Karahalios A, Brown KA, Gunter MJ, Milne RL, English DR, Lynch BM. Sex-steroid hormones and risk of postmenopausal estrogen receptor-positive breast cancer: a case-cohort analysis. RESEARCH SQUARE 2023:rs.3.rs-3406466. [PMID: 37886482 PMCID: PMC10602098 DOI: 10.21203/rs.3.rs-3406466/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Purpose Sex-steroid hormones are associated with postmenopausal breast cancer but potential confounding from other biological pathways is rarely considered. We estimated risk ratios for sex-steroid hormone biomarkers in relation to postmenopausal estrogen receptor (ER)-positive breast cancer, while accounting for biomarkers from insulin/insulin-like growth factor-signaling and inflammatory pathways. Methods This analysis included 1,208 women from a case-cohort study of postmenopausal breast cancer within the Melbourne Collaborative Cohort Study. Weighted Poisson regression with a robust variance estimator was used to estimate risk ratios (RRs) and 95% confidence intervals (CIs) of postmenopausal ER-positive breast cancer, per doubling plasma concentration of progesterone, estrogens, androgens, and sex hormone binding globulin (SHBG). Analyses included sociodemographic and lifestyle confounders, and other biomarkers identified as potential confounders. Results Increased risks of postmenopausal ER-positive breast cancer were observed per doubling plasma concentration of progesterone (RR: 1.22, 95% CI: 1.03 to 1.44), androstenedione (RR: 1.20, 95% CI: 0.99 to 1.45), dehydroepiandrosterone (RR: 1.15, 95% CI: 1.00 to 1.34), total testosterone (RR: 1.11, 95% CI: 0.96 to 1.29), free testosterone (RR: 1.12, 95% CI: 0.98 to 1.28), estrone (RR: 1.21, 95% CI: 0.99 to 1.48), total estradiol (RR: 1.19, 95% CI: 1.02 to 1.39) and free estradiol (RR: 1.22, 95% CI: 1.05 to 1.41). A possible decreased risk was observed for SHBG (RR: 0.83, 95% CI: 0.66 to 1.05). Conclusion Progesterone, estrogens and androgens likely increase postmenopausal ER-positive breast cancer risk, whereas SHBG may decrease risk. These findings strengthen the causal evidence surrounding the sex hormone-driven nature of postmenopausal breast cancer.
Collapse
|
44
|
Kong X, Yan W, Sun W, Zhang Y, Yang HJ, Chen M, Chen H, de Vere White RW, Zhang J, Chen X. Isoform-specific disruption of the TP73 gene reveals a critical role for TAp73γ in tumorigenesis via leptin. eLife 2023; 12:e82115. [PMID: 37650871 PMCID: PMC10471163 DOI: 10.7554/elife.82115] [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: 07/23/2022] [Accepted: 08/01/2023] [Indexed: 09/01/2023] Open
Abstract
TP73, a member of the p53 family, is expressed as TAp73 and ΔNp73 along with multiple C-terminal isoforms (α-η). ΔNp73 is primarily expressed in neuronal cells and necessary for neuronal development. Interestingly, while TAp73α is a tumor suppressor and predominantly expressed in normal cells, TAp73 is found to be frequently altered in human cancers, suggesting a role of TAp73 C-terminal isoforms in tumorigenesis. To test this, the TCGA SpliceSeq database was searched and showed that exon 11 (E11) exclusion occurs frequently in several human cancers. We also found that p73α to p73γ isoform switch resulting from E11 skipping occurs frequently in human prostate cancers and dog lymphomas. To determine whether p73α to p73γ isoform switch plays a role in tumorigenesis, CRISPR technology was used to generate multiple cancer cell lines and a mouse model in that Trp73 E11 is deleted. Surprisingly, we found that in E11-deificient cells, p73γ becomes the predominant isoform and exerts oncogenic activities by promoting cell proliferation and migration. In line with this, E11-deficient mice were more prone to obesity and B-cell lymphomas, indicating a unique role of p73γ in lipid metabolism and tumorigenesis. Additionally, we found that E11-deficient mice phenocopies Trp73-deficient mice with short lifespan, infertility, and chronic inflammation. Mechanistically, we showed that Leptin, a pleiotropic adipocytokine involved in energy metabolism and oncogenesis, was highly induced by p73γ,necessary for p73γ-mediated oncogenic activity, and associated with p73α to γ isoform switch in human prostate cancer and dog lymphoma. Finally, we showed that E11-knockout promoted, whereas knockdown of p73γ or Leptin suppressed, xenograft growth in mice. Our study indicates that the p73γ-Leptin pathway promotes tumorigenesis and alters lipid metabolism, which may be targeted for cancer management.
Collapse
Affiliation(s)
- Xiangmudong Kong
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, DavisDavisUnited States
| | - Wensheng Yan
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, DavisDavisUnited States
| | - Wenqiang Sun
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, DavisDavisUnited States
| | - Yanhong Zhang
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, DavisDavisUnited States
| | - Hee Jung Yang
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, DavisDavisUnited States
| | - Mingyi Chen
- Department of Pathology, University of Texas Southwestern Medical CenterDallasUnited States
| | - Hongwu Chen
- Department of Biochemistry and Molecular Medicine, University of California, DavisDavisUnited States
| | - Ralph W de Vere White
- Department of Urology Surgery, School of Medicine, University of California, DavisDavisUnited States
| | - Jin Zhang
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, DavisDavisUnited States
| | - Xinbin Chen
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, DavisDavisUnited States
| |
Collapse
|
45
|
Bocian-Jastrzębska A, Malczewska-Herman A, Kos-Kudła B. Role of Leptin and Adiponectin in Carcinogenesis. Cancers (Basel) 2023; 15:4250. [PMID: 37686525 PMCID: PMC10486522 DOI: 10.3390/cancers15174250] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/18/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023] Open
Abstract
Hormones produced by adipocytes, leptin and adiponectin, are associated with the process of carcinogenesis. Both of these adipokines have well-proven oncologic potential and can affect many aspects of tumorigenesis, from initiation and primary tumor growth to metastatic progression. Involvement in the formation of cancer includes interactions with the tumor microenvironment and its components, such as tumor-associated macrophages, cancer-associated fibroblasts, extracellular matrix and matrix metalloproteinases. Furthermore, these adipokines participate in the epithelial-mesenchymal transition and connect to angiogenesis, which is critical for cancer invasiveness and cancer cell migration. In addition, an enormous amount of evidence has demonstrated that altered concentrations of these adipocyte-derived hormones and the expression of their receptors in tumors are associated with poor prognosis in various types of cancer. Therefore, leptin and adiponectin dysfunction play a prominent role in cancer and impact tumor invasion and metastasis in different ways. This review clearly and comprehensively summarizes the recent findings and presents the role of leptin and adiponectin in cancer initiation, promotion and progression, focusing on associations with the tumor microenvironment and its components as well as roles in the epithelial-mesenchymal transition and angiogenesis.
Collapse
Affiliation(s)
- Agnes Bocian-Jastrzębska
- Department of Endocrinology and Neuroendocrine Tumors, Department of Pathophysiology and Endocrinogy, Medical University of Silesia, 40-514 Katowice, Poland; (A.M.-H.); (B.K.-K.)
| | | | | |
Collapse
|
46
|
Glassman I, Le N, Asif A, Goulding A, Alcantara CA, Vu A, Chorbajian A, Mirhosseini M, Singh M, Venketaraman V. The Role of Obesity in Breast Cancer Pathogenesis. Cells 2023; 12:2061. [PMID: 37626871 PMCID: PMC10453206 DOI: 10.3390/cells12162061] [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: 05/22/2023] [Revised: 08/03/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
Research has shown that obesity increases the risk for type 2 diabetes mellitus (Type 2 DM) by promoting insulin resistance, increases serum estrogen levels by the upregulation of aromatase, and promotes the release of reactive oxygen species (ROS) by macrophages. Increased circulating glucose has been shown to activate mammalian target of rapamycin (mTOR), a significant signaling pathway in breast cancer pathogenesis. Estrogen plays an instrumental role in estrogen-receptor-positive breast cancers. The role of ROS in breast cancer warrants continued investigation, in relation to both pathogenesis and treatment of breast cancer. We aim to review the role of obesity in breast cancer pathogenesis and novel therapies mediating obesity-associated breast cancer development. We explore the association between body mass index (BMI) and breast cancer incidence and the mechanisms by which oxidative stress modulates breast cancer pathogenesis. We discuss the role of glutathione, a ubiquitous antioxidant, in breast cancer therapy. Lastly, we review breast cancer therapies targeting mTOR signaling, leptin signaling, blood sugar reduction, and novel immunotherapy targets.
Collapse
Affiliation(s)
- Ira Glassman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA (N.L.); (A.A.); (C.A.A.); (M.M.)
| | - Nghia Le
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA (N.L.); (A.A.); (C.A.A.); (M.M.)
| | - Aamna Asif
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA (N.L.); (A.A.); (C.A.A.); (M.M.)
| | - Anabel Goulding
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA (N.L.); (A.A.); (C.A.A.); (M.M.)
| | - Cheldon Ann Alcantara
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA (N.L.); (A.A.); (C.A.A.); (M.M.)
| | - Annie Vu
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA (N.L.); (A.A.); (C.A.A.); (M.M.)
| | - Abraham Chorbajian
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA (N.L.); (A.A.); (C.A.A.); (M.M.)
| | - Mercedeh Mirhosseini
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA (N.L.); (A.A.); (C.A.A.); (M.M.)
| | - Manpreet Singh
- Corona Regional Medical Center, Department of Emergency Medicine, Corona, CA 92882, USA
| | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA (N.L.); (A.A.); (C.A.A.); (M.M.)
| |
Collapse
|
47
|
Zhao Y, Jia H, Hua X, An T, Song J. Cardio-oncology: Shared Genetic, Metabolic, and Pharmacologic Mechanism. Curr Cardiol Rep 2023; 25:863-878. [PMID: 37493874 PMCID: PMC10403418 DOI: 10.1007/s11886-023-01906-6] [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] [Accepted: 06/11/2023] [Indexed: 07/27/2023]
Abstract
PURPOSE OF REVIEW The article aims to investigate the complex relationship between cancer and cardiovascular disease (CVD), with a focus on the effects of cancer treatment on cardiac health. RECENT FINDINGS Advances in cancer treatment have improved long-term survival rates, but CVD has emerged as a leading cause of morbidity and mortality in cancer patients. The interplay between cancer itself, treatment methods, homeostatic changes, and lifestyle modifications contributes to this comorbidity. Recent research in the field of cardio-oncology has revealed common genetic mutations, risk factors, and metabolic features associated with the co-occurrence of cancer and CVD. This article provides a comprehensive review of the latest research in cardio-oncology, including common genetic mutations, risk factors, and metabolic features, and explores the interactions between cancer treatment and CVD drugs, proposing novel approaches for the management of cancer and CVD.
Collapse
Affiliation(s)
- Yiqi Zhao
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, 100037 Beijing, China
| | - Hao Jia
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, 100037 Beijing, China
| | - Xiumeng Hua
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, 100037 Beijing, China
| | - Tao An
- Department of Cardiology, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiangping Song
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, 100037 Beijing, China
| |
Collapse
|
48
|
Lee-Rueckert M, Canyelles M, Tondo M, Rotllan N, Kovanen PT, Llorente-Cortes V, Escolà-Gil JC. Obesity-induced changes in cancer cells and their microenvironment: Mechanisms and therapeutic perspectives to manage dysregulated lipid metabolism. Semin Cancer Biol 2023; 93:36-51. [PMID: 37156344 DOI: 10.1016/j.semcancer.2023.05.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 04/05/2023] [Accepted: 05/05/2023] [Indexed: 05/10/2023]
Abstract
Obesity has been closely related to cancer progression, recurrence, metastasis, and treatment resistance. We aim to review recent progress in the knowledge on the obese macroenvironment and the generated adipose tumor microenvironment (TME) inducing lipid metabolic dysregulation and their influence on carcinogenic processes. Visceral white adipose tissue expansion during obesity exerts systemic or macroenvironmental effects on tumor initiation, growth, and invasion by promoting inflammation, hyperinsulinemia, growth-factor release, and dyslipidemia. The dynamic relationship between cancer and stromal cells of the obese adipose TME is critical for cancer cell survival and proliferation as well. Experimental evidence shows that secreted paracrine signals from cancer cells can induce lipolysis in cancer-associated adipocytes, causing them to release free fatty acids and acquire a fibroblast-like phenotype. Such adipocyte delipidation and phenotypic change is accompanied by an increased secretion of cytokines by cancer-associated adipocytes and tumor-associated macrophages in the TME. Mechanistically, the availability of adipose TME free fatty acids and tumorigenic cytokines concomitant with the activation of angiogenic processes creates an environment that favors a shift in the cancer cells toward an aggressive phenotype associated with increased invasiveness. We conclude that restoring the aberrant metabolic alterations in the host macroenvironment and in adipose TME of obese subjects would be a therapeutic option to prevent cancer development. Several dietary, lipid-based, and oral antidiabetic pharmacological therapies could potentially prevent tumorigenic processes associated with the dysregulated lipid metabolism closely linked to obesity.
Collapse
Affiliation(s)
| | - Marina Canyelles
- Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Mireia Tondo
- Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Noemi Rotllan
- Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | | | - Vicenta Llorente-Cortes
- Wihuri Research Institute, Helsinki, Finland; Institute of Biomedical Research of Barcelona (IIBB)-Spanish National Research Council (CSIC), Barcelona, Spain; CIBERCV, Institute of Health Carlos III, 28029 Madrid, Spain.
| | - Joan Carles Escolà-Gil
- Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain.
| |
Collapse
|
49
|
Lathigara D, Kaushal D, Wilson RB. Molecular Mechanisms of Western Diet-Induced Obesity and Obesity-Related Carcinogenesis-A Narrative Review. Metabolites 2023; 13:metabo13050675. [PMID: 37233716 DOI: 10.3390/metabo13050675] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 05/05/2023] [Accepted: 05/18/2023] [Indexed: 05/27/2023] Open
Abstract
The present study aims to provide a narrative review of the molecular mechanisms of Western diet-induced obesity and obesity-related carcinogenesis. A literature search of the Cochrane Library, Embase and Pubmed databases, Google Scholar and the grey literature was conducted. Most of the molecular mechanisms that induce obesity are also involved in the twelve Hallmarks of Cancer, with the fundamental process being the consumption of a highly processed, energy-dense diet and the deposition of fat in white adipose tissue and the liver. The generation of crown-like structures, with macrophages surrounding senescent or necrotic adipocytes or hepatocytes, leads to a perpetual state of chronic inflammation, oxidative stress, hyperinsulinaemia, aromatase activity, activation of oncogenic pathways and loss of normal homeostasis. Metabolic reprogramming, epithelial mesenchymal transition, HIF-1α signalling, angiogenesis and loss of normal host immune-surveillance are particularly important. Obesity-associated carcinogenesis is closely related to metabolic syndrome, hypoxia, visceral adipose tissue dysfunction, oestrogen synthesis and detrimental cytokine, adipokine and exosomal miRNA release. This is particularly important in the pathogenesis of oestrogen-sensitive cancers, including breast, endometrial, ovarian and thyroid cancer, but also 'non-hormonal' obesity-associated cancers such as cardio-oesophageal, colorectal, renal, pancreatic, gallbladder and hepatocellular adenocarcinoma. Effective weight loss interventions may improve the future incidence of overall and obesity-associated cancer.
Collapse
Affiliation(s)
- Dhruvi Lathigara
- Department General Surgery, UWS, Campbelltown Hospital, Campbelltown, NSW 2560, Australia
| | - Devesh Kaushal
- Department General Surgery, UWS, Campbelltown Hospital, Campbelltown, NSW 2560, Australia
| | - Robert Beaumont Wilson
- Department Upper Gastrointestinal Surgery, UNSW, Liverpool Hospital, Liverpool, NSW 2170, Australia
| |
Collapse
|
50
|
Clemente-Suárez VJ, Redondo-Flórez L, Beltrán-Velasco AI, Martín-Rodríguez A, Martínez-Guardado I, Navarro-Jiménez E, Laborde-Cárdenas CC, Tornero-Aguilera JF. The Role of Adipokines in Health and Disease. Biomedicines 2023; 11:biomedicines11051290. [PMID: 37238961 DOI: 10.3390/biomedicines11051290] [Citation(s) in RCA: 104] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/21/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
Adipokines are cell-signaling proteins secreted by adipose tissue that has been related to a low-grade state of inflammation and different pathologies. The present review aims to analyze the role of adipokines in health and disease in order to understand the important functions and effects of these cytokines. For this aim, the present review delves into the type of adipocytes and the cytokines produced, as well as their functions; the relations of adipokines in inflammation and different diseases such as cardiovascular, atherosclerosis, mental diseases, metabolic disorders, cancer, and eating behaviors; and finally, the role of microbiota, nutrition, and physical activity in adipokines is discussed. This information would allow for a better understanding of these important cytokines and their effects on body organisms.
Collapse
Affiliation(s)
| | - Laura Redondo-Flórez
- Department of Health Sciences, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, C/Tajo s/n, 28670 Madrid, Spain
| | - Ana Isabel Beltrán-Velasco
- Department of Psychology, Faculty of Life and Natural Sciences, University of Nebrija, C/del Hostal, 28248 Madrid, Spain
| | | | - Ismael Martínez-Guardado
- BRABE Group, Department of Psychology, Faculty of Life and Natural Sciences, University of Nebrija, C/del Hostal, 28248 Madrid, Spain
| | | | | | | |
Collapse
|