1
|
McCaffrey D, Weickert CS, Walker AK. Blood IL-1α and IL-6 predict specific breast cancer-induced increases in hippocampal pro-inflammatory cytokines in mice. Cytokine 2025; 186:156826. [PMID: 39667084 DOI: 10.1016/j.cyto.2024.156826] [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: 04/23/2024] [Revised: 11/22/2024] [Accepted: 12/02/2024] [Indexed: 12/14/2024]
Abstract
Neuroinflammation is a key factor in cognitive and behavioral changes seen in patients with non-CNS cancers, and cytokine levels in the blood are often used as a proxy for brain inflammation. However, this approach has yielded inconsistent results, and a common inflammatory signature remains elusive. To explore whether a blood-to-brain inflammatory signature exists across breast cancer types, we assessed cytokine and glial protein responses in the hippocampus, prefrontal cortex (PFC), and their relationship to serum cytokines in mice bearing three different mammary cancers (n = 40). While cytokine profiles in both serum and brain varied by cancer type, IL-1β and IL-4 were consistently altered across brain regions. In some cases, elevated serum IL-1α and IL-6 correlated with increased hippocampal IL-6. These findings support the use of blood cytokines to identify cancer patients at risk for cognitive and psychiatric comorbidities. However, our data also suggest that relying solely on serum cytokines may lead to under-diagnosis, as some mice exhibited brain cytokine elevations without changes in serum levels. This underscores the need for a broader range of inflammatory markers in blood to better identify at-risk patients. Brain region-specific differences in the cytokine response to mammary cancer highlighted the hippocampus as more vulnerable to cancer-induced inflammation than the PFC. We observed region-specific glial cell reactivity, however, only astrocyte and oligodendrocyte markers were correlated with cytokine changes within the hippocampus. Elevated serum IL-1α and IL-6 were correlated with reduced cortical astrocyte reactivity, suggesting that these cytokines can inform glial cell-specific changes in this region.
Collapse
Affiliation(s)
- Delyse McCaffrey
- Laboratory of ImmunoPsychiatry, Neuroscience Research Australia, Randwick, New South Wales, Australia; Discipline of Psychiatry and Mental Health, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Cynthia Shannon Weickert
- Discipline of Psychiatry and Mental Health, Faculty of Medicine, University of New South Wales, Sydney, Australia; Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, New South Wales, Australia; Department of Neuroscience & Physiology, Upstate Medical University, Syracuse, NY, USA
| | - Adam K Walker
- Laboratory of ImmunoPsychiatry, Neuroscience Research Australia, Randwick, New South Wales, Australia; Discipline of Psychiatry and Mental Health, Faculty of Medicine, University of New South Wales, Sydney, Australia; Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia.
| |
Collapse
|
2
|
Satam S, Palekar N, Premkumar K, Shankar BS. Sirtinol, a SIRT1 inhibitor, inhibits the EMT and metastasis of 4T1 breast cancer cells and impacts the tumor microenvironment. Immunopharmacol Immunotoxicol 2024; 46:829-842. [PMID: 39373058 DOI: 10.1080/08923973.2024.2412110] [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: 08/19/2023] [Accepted: 09/28/2024] [Indexed: 10/08/2024]
Abstract
INTRODUCTION The impact of epigenetic drugs on metastasis and the immunological microenvironment is poorly understood. In this study, we looked at how sirtinol, a SIRT1 inhibitor, affected epithelial-mesenchymal transition (EMT), metastasis, and the immune cells. MATERIALS AND METHODS In vitro experiments were carried out using tumor conditioned medium (TCM). For in vivo experiments, sirtinol was administered i.p. in tumor bearing BALB/c mice at a dose of 2 mg/kg body weight either alone or in combination with cisplatin. Estimation of cytokines was carried out using ELISA or ELIspot. Estimation of different markers was done using flow cytometry or western blot. RESULTS Sirtinol, a SIRT1 inhibitor, was found to be cytotoxic to 4T1 breast cancer cells with no synergistic effects with cisplatin, both under in vitro and in vivo conditions (p < 0.05). Sirtinol significantly reduced cancer cell metastasis to the spleen which was supported by in vitro findings such as decreased vimentin expression and cell mobility in migration and wound healing assays (p < 0.01). Studies on the effects of 4T1 tumor-conditioned medium on spleen cells indicated changes in T cell proliferation as well as differentiation (p < 0.01). In tumor bearing mice, spleen cells showed elevated IFN-γ secretion, increased CD11b+ cells, and decreased T cells (p < 0.01). This was reversed by sirtinol as well as the combination treatment, which may also have contributed to metastasis inhibition (p < 0.01). CONCLUSION Sirtinol, a SIRT1 inhibitor inhibits EMT and metastasis of 4T1 breast cancer cells and also has an impact on the immune microenvironment.
Collapse
Affiliation(s)
- Sharvari Satam
- Immunology Section, Radiation Biology & Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Mumbai, India
| | - Nitya Palekar
- Immunology Section, Radiation Biology & Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Mumbai, India
| | - Kavitha Premkumar
- Immunology Section, Radiation Biology & Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Mumbai, India
| | - Bhavani S Shankar
- Immunology Section, Radiation Biology & Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
3
|
Xu H, Sun Z, Wang G, Li R. The Impact of Depression on Detrimental Changes in Bone Microstructure in Female Mice. Neuropsychiatr Dis Treat 2024; 20:1421-1433. [PMID: 39049938 PMCID: PMC11268775 DOI: 10.2147/ndt.s454865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 07/02/2024] [Indexed: 07/27/2024] Open
Abstract
Background Several clinical studies have examined the connection between depression and bone loss, but the cause-and-effect relationship between the two conditions, especially in animal models, is not well-studied. Methods A total of 32 female mice were, randomly divided into control group (CON, n=19) and depression group (DEP, n=13). The mice in the DEP group were subjected to 21 consecutive days of restraint stress, following depressive-like behaviors were assessment. The femurs were collected using Micro-Computed Tomography (μCT) and histochemical staining. In parallel, levels of serotonin-related proteins in the brain were measured using Western blot analysis, and sex hormone profiles were determined through liquid chromatography-mass spectrometry/mass spectrometry (LC-MS/MS). Results The mice in the DEP group exhibited clear signs of depressive-like behaviors and an increase in serotonin transporter levels (t=-2.435, P< 0.05). In comparison to the CON mice, the DEP mice showed a decrease in bone mineral density (t =3.741, P< 0.05), bone surface area density (t =8.009, P<0.01), percent bone volume (t =4.293, P< 0.05), trabecular number (t =5.844, P<0.01), and connected density (t =11.000, P< 0.05). Additionally, there was an increase in trabecular separation (t =-7.436, P<0.01) in DEP mice. Furthermore, the DEP mice displayed a significant reduction in serum estrogen levels (t =4.340, P< 0.05) and changes in its metabolite (t =-3.325, P< 0.05), while the levels of androgens remained unchanged. Conclusion The restraint stress not only led to the development of depressive-like behaviors but also disrupted the estrogen metabolism pathway, resulting in damage to bone mass and microstructure in female mice. These findings suggest that stress-induced depression may pose a risk for bone loss in female mice by altering estrogen metabolism pathways.
Collapse
Affiliation(s)
- Hong Xu
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Zuoli Sun
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Gang Wang
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, People’s Republic of China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, People’s Republic of China
| | - Rena Li
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, People’s Republic of China
| |
Collapse
|
4
|
Gonçalves IV, Pinheiro-Rosa N, Torres L, Oliveira MDA, Rapozo Guimarães G, Leite CDS, Ortega JM, Lopes MTP, Faria AMC, Martins MLB, Felicori LF. Dynamic changes in B cell subpopulations in response to triple-negative breast cancer development. Sci Rep 2024; 14:11576. [PMID: 38773133 PMCID: PMC11109097 DOI: 10.1038/s41598-024-60243-y] [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: 09/26/2023] [Accepted: 04/19/2024] [Indexed: 05/23/2024] Open
Abstract
Despite presenting a worse prognosis and being associated with highly aggressive tumors, triple-negative breast cancer (TNBC) is characterized by the higher frequency of tumor-infiltrating lymphocytes, which have been implicated in better overall survival and response to therapy. Though recent studies have reported the capacity of B lymphocytes to recognize overly-expressed normal proteins, and tumor-associated antigens, how tumor development potentially modifies B cell response is yet to be elucidated. Our findings reveal distinct effects of 4T1 and E0771 murine tumor development on B cells in secondary lymphoid organs. Notably, we observe a significant expansion of total B cells and plasma cells in the tumor-draining lymph nodes (tDLNs) as early as 7 days after tumor challenge in both murine models, whereas changes in the spleen are less pronounced. Surprisingly, within the tumor microenvironment (TME) of both models, we detect distinct B cell subpopulations, but tumor development does not appear to cause major alterations in their frequency over time. Furthermore, our investigation into B cell regulatory phenotypes highlights that the B10 Breg phenotype remains unaffected in the evaluated tissues. Most importantly, we identified an increase in CD19 + LAG-3 + cells in tDLNs of both murine models. Interestingly, although CD19 + LAG-3 + cells represent a minor subset of total B cells (< 3%) in all evaluated tissues, most of these cells exhibit elevated expression of IgD, suggesting that LAG-3 may serve as an activation marker for B cells. Corroborating with these findings, we detected distinct cell cycle and proliferation genes alongside LAG-3 analyzing scRNA-Seq data from a cohort of TNBC patients. More importantly, our study suggests that the presence of LAG-3 B cells in breast tumors could be associated with a good prognosis, as patients with higher levels of LAG-3 B cell transcripts had a longer progression-free interval (PFI). This novel insight could pave the way for targeted therapies that harness the unique properties of LAG-3 + B cells, potentially offering new avenues for improving patient outcomes in TNBC. Further research is warranted to unravel the mechanistic pathways of these cells and to validate their prognostic value in larger, diverse patient cohorts.
Collapse
Affiliation(s)
- Igor Visconte Gonçalves
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627 - Pampulha, Belo Horizonte, MG, 31270-901, Brazil
| | - Natália Pinheiro-Rosa
- NYU Grossman School of Medicine, NYU Langone Health, New York University, 550 1st Ave, New York, NY, 10016, USA
| | - Lícia Torres
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627 - Pampulha, Belo Horizonte, MG, 31270-901, Brazil
| | - Mariana de Almeida Oliveira
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627 - Pampulha, Belo Horizonte, MG, 31270-901, Brazil
| | - Gabriela Rapozo Guimarães
- Instituto Nacional de Câncer, Ministério da Saúde, Coordenação de Pesquisa, Laboratório de Bioinformática e Biologia Computacional - Rua André Cavalcanti, 37, 1 Andar, Centro, Rio de Janeiro, RJ, 20231050, Brasil
| | - Christiana da Silva Leite
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627 - Pampulha, Belo Horizonte, MG, 31270-901, Brazil
| | - José Miguel Ortega
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627 - Pampulha, Belo Horizonte, MG, 31270-901, Brazil
| | - Miriam Teresa Paz Lopes
- Department of Pharmacology, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627 - Pampulha, Belo Horizonte, MG, 31270-901, Brazil
| | - Ana Maria Caetano Faria
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627 - Pampulha, Belo Horizonte, MG, 31270-901, Brazil
| | - Mariana Lima Boroni Martins
- Instituto Nacional de Câncer, Ministério da Saúde, Coordenação de Pesquisa, Laboratório de Bioinformática e Biologia Computacional - Rua André Cavalcanti, 37, 1 Andar, Centro, Rio de Janeiro, RJ, 20231050, Brasil
| | - Liza Figueiredo Felicori
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627 - Pampulha, Belo Horizonte, MG, 31270-901, Brazil.
| |
Collapse
|
5
|
Monterroza L, Parrilla MM, Samaranayake SG, Rivera-Rodriguez DE, Yoon SB, Bommireddy R, Hosten J, Barragan LC, Marcus A, Dobosh BS, Selvaraj P, Tirouvanziam R. Tumor-Intrinsic Enhancer of Zeste Homolog 2 Controls Immune Cell Infiltration, Tumor Growth, and Lung Metastasis in a Triple-Negative Breast Cancer Model. Int J Mol Sci 2024; 25:5392. [PMID: 38791429 PMCID: PMC11121204 DOI: 10.3390/ijms25105392] [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: 04/02/2024] [Revised: 05/06/2024] [Accepted: 05/12/2024] [Indexed: 05/26/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive and highly metastatic type of tumor. TNBC is often enriched in tumor-infiltrating neutrophils (TINs), which support cancer growth in part by counteracting tumor-infiltrating lymphocytes (TILs). Prior studies identified the enhancer of zeste homolog 2 (EZH2) as a pro-tumor methyltransferase in primary and metastatic TNBCs. We hypothesized that EZH2 inhibition in TNBC cells per se would exert antitumor activity by altering the tumor immune microenvironment. To test this hypothesis, we used CRISPR to generate EZH2 gene knockout (KO) and overexpressing (OE) lines from parent (wild-type-WT) 4T1 cells, an established murine TNBC model, resulting in EZH2 protein KO and OE, respectively. In vitro, EZH2 KO and OE cells showed early, transient changes in replicative capacity and invasiveness, and marked changes in surface marker profile and cytokine/chemokine secretion compared to WT cells. In vivo, EZH2 KO cells showed significantly reduced primary tumor growth and a 10-fold decrease in lung metastasis compared to WT cells, while EZH2 OE cells were unchanged. Compared to WT tumors, TIN:TIL ratios were greatly reduced in EZH2 KO tumors but unchanged in EZH2 OE tumors. Thus, EZH2 is key to 4T1 aggressiveness as its tumor-intrinsic knockout alters their in vitro secretome and in vivo primary tumor growth, TIN/TIL poise, and metastasis.
Collapse
Affiliation(s)
- Lenore Monterroza
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (L.M.); (M.M.P.); (J.H.); (B.S.D.)
- Department of Pathology & Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA;
| | - Maria M. Parrilla
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (L.M.); (M.M.P.); (J.H.); (B.S.D.)
| | | | - Dormarie E. Rivera-Rodriguez
- Department of Microbiology & Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA; (D.E.R.-R.); (L.C.B.)
| | - Sung Bo Yoon
- Department of Hematology & Medical Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA; (S.B.Y.); (A.M.)
| | - Ramireddy Bommireddy
- Department of Pathology & Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA;
| | - Justin Hosten
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (L.M.); (M.M.P.); (J.H.); (B.S.D.)
| | - Luisa Cervantes Barragan
- Department of Microbiology & Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA; (D.E.R.-R.); (L.C.B.)
| | - Adam Marcus
- Department of Hematology & Medical Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA; (S.B.Y.); (A.M.)
| | - Brian S. Dobosh
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (L.M.); (M.M.P.); (J.H.); (B.S.D.)
| | - Periasamy Selvaraj
- Department of Pathology & Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA;
| | - Rabindra Tirouvanziam
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (L.M.); (M.M.P.); (J.H.); (B.S.D.)
| |
Collapse
|
6
|
Strehle LD, Otto-Dobos LD, Grant CV, Glasper ER, Pyter LM. Microglia contribute to mammary tumor-induced neuroinflammation in a female mouse model. FASEB J 2024; 38:e23419. [PMID: 38236370 PMCID: PMC10832463 DOI: 10.1096/fj.202301580rr] [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: 08/03/2023] [Revised: 11/13/2023] [Accepted: 12/27/2023] [Indexed: 01/19/2024]
Abstract
Following diagnosis but before treatment, up to 30% of breast cancer patients report behavioral side effects (e.g., anxiety, depression, memory impairment). Our rodent mammary tumor model recapitulates aspects of these behavioral sequelae, as well as elevated circulating and brain inflammatory mediators. Neuroinflammation is a proposed mechanism underlying the etiology of mood disorders and cognitive deficits, and therefore may be contributing to tumor-associated behavioral side effects. The cellular mechanisms by which tumor-induced neuroinflammation occurs remain unknown, making targeted treatment approaches inaccessible. Here, we tested the hypotheses that microglia are the primary cells driving tumor-induced neuroinflammation and behavioral side effects. Young adult female BALB/c mice were induced with a 67NR mammary tumor; tumor-free controls underwent a sham surgery. Mammary tumors increased IBA1+ and GFAP+ staining in the amygdala and hippocampus relative to tumor-free controls. However, tumors did not alter gene expression of Percoll-enriched microglia isolated from the whole brain. While cognitive, social, and anhedonia-like behaviors were not altered in tumor-bearing mice, tumors increased central tendency in the open-field test; microglia depletion did not reverse this effect. Brain region RT-qPCR data indicated that microglia depletion attenuated tumor-induced elevations of neuroinflammatory gene expression in a region- and mediator-specific manner. These results indicate a causal role of microglia in tumor-induced neuroinflammation. This research advances our understanding of the cellular mechanisms underlying tumor-induced neuroinflammation in order to understand how brain responses (e.g., behavior) may be altered with subsequent cancer-related immune challenges.
Collapse
Affiliation(s)
- Lindsay D. Strehle
- Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Lauren D. Otto-Dobos
- Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Corena V. Grant
- Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Erica R. Glasper
- Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Neuroscience, Ohio State University, Columbus, OH, USA
| | - Leah M. Pyter
- Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Psychiatry and Behavioral Health, Ohio State University, Columbus, OH, USA
- Department of Neuroscience, Ohio State University, Columbus, OH, USA
| |
Collapse
|
7
|
Orbach SM, DeVaull CY, Bealer EJ, Ross BC, Jeruss JS, Shea LD. An engineered niche delineates metastatic potential of breast cancer. Bioeng Transl Med 2024; 9:e10606. [PMID: 38193115 PMCID: PMC10771563 DOI: 10.1002/btm2.10606] [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: 05/22/2023] [Revised: 08/29/2023] [Accepted: 09/20/2023] [Indexed: 01/10/2024] Open
Abstract
Metastatic breast cancer is often not diagnosed until secondary tumors have become macroscopically visible and millions of tumor cells have invaded distant tissues. Yet, metastasis is initiated by a cascade of events leading to formation of the pre-metastatic niche, which can precede tumor formation by a matter of years. We aimed to distinguish the potential for metastatic disease from nonmetastatic disease at early times in triple-negative breast cancer using sister cell lines 4T1 (metastatic), 4T07 (invasive, nonmetastatic), and 67NR (nonmetastatic). We used a porous, polycaprolactone scaffold, that serves as an engineered metastatic niche, to identify metastatic disease through the characteristics of the microenvironment. Analysis of the immune cell composition at the scaffold was able to distinguish noninvasive 67NR tumor-bearing mice from 4T07 and 4T1 tumor-bearing mice but could not delineate metastatic potential between the two invasive cell lines. Gene expression in the scaffolds correlated with the up-regulation of cancer hallmarks (e.g., angiogenesis, hypoxia) in the 4T1 mice relative to 4T07 mice. We developed a 9-gene signature (Dhx9, Dusp12, Fth1, Ifitm1, Ndufs1, Pja2, Slc1a3, Soga1, Spon2) that successfully distinguished 4T1 disease from 67NR or 4T07 disease throughout metastatic progression. Furthermore, this signature proved highly effective at distinguishing diseased lungs in publicly available datasets of mouse models of metastatic breast cancer and in human models of lung cancer. The early and accurate detection of metastatic disease that could lead to early treatment has the potential to improve patient outcomes and quality of life.
Collapse
Affiliation(s)
- Sophia M. Orbach
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | | | - Elizabeth J. Bealer
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Brian C. Ross
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Jacqueline S. Jeruss
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
- Department of PathologyUniversity of MichiganAnn ArborMichiganUSA
- Department of SurgeryUniversity of MichiganAnn ArborMichiganUSA
| | - Lonnie D. Shea
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
- Department of Chemical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
8
|
Ramos JP, Abdel-Salam MAL, Nobre DAB, Glanzmann N, de Souza CP, Leite EA, de Abreu Teles PP, Barbosa AS, Barcelos LS, Dos Reis DC, Cassali GD, de Lima ME, de Castro QJT, Grabe-Guimarães A, da Silva AD, de Souza-Fagundes EM. Acute toxicity and antitumor potential of 1,3,4-trisubstituted-1,2,3-triazole dhmtAc-loaded liposomes on a triple-negative breast cancer model. Arch Pharm (Weinheim) 2022; 355:e2200004. [PMID: 35621705 DOI: 10.1002/ardp.202200004] [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: 01/11/2022] [Revised: 04/28/2022] [Accepted: 05/02/2022] [Indexed: 11/05/2022]
Abstract
For the first time, compounds developed from the 1,2,3-triazole scaffold were evaluated as novel drugs to treat triple-negative breast cancer (TNBC). Four organic salts were idealized as nonclassical bioisosteres of miltefosine, which is used in the topical treatment for skin metastasizing breast carcinoma. Among them, derivative dhmtAc displayed better solubility and higher cytotoxicity against the human breast adenocarcinoma cell line and mouse 4T1 cell lines, which are representatives of TNBC. In vitro assays revealed that dhmtAc interferes with cell integrity, confirmed by lactate dehydogenase leakage. Due to its human peripheral blood mononuclear cell (PBMC) toxicity, dhmtAc in vivo studies were carried out with the drug incorporated in a long-circulating and pH-sensitive liposome (SpHL-dhmtAc), and the acute toxicity in BALB/c mice was determined. Free dhmtAc displayed cardiac and pulmonary toxicity after the systemic administration of 5 mg/kg doses. On the other hand, SpHL-dhmtAc displayed no toxicity at 20 mg/kg. The in vivo antitumor effect of SpHL-dhmtAc was investigated using the 4T1 heterotopic murine model. Intravenous administration of SpHL-dhmtAc reduced the tumor volume and weight, without interfering with the body weight, compared with the control group and the dhmtAc free form. The incorporation of the triazole compound in the liposome allowed the demonstration of its anticancer potential. These findings evidenced 1,3,4-trisubstituted-1,2,3-triazole as a promising scaffold for the development of novel drugs with applicability for the treatment of patients with TNBC.
Collapse
Affiliation(s)
- Jonas P Ramos
- Departamento de Fisiologia e Biofísica, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mostafa A L Abdel-Salam
- Departamento de Fisiologia e Biofísica, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Daniel A B Nobre
- Departamento de Fisiologia e Biofísica, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Nicolas Glanzmann
- Departamento de Química, Universidade Federal de Juiz de Fora, Juiz de Fora, Brazil
| | - Camila P de Souza
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Elaine A Leite
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Pedro P de Abreu Teles
- Departamento de Patologia Geral, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Alan S Barbosa
- Departamento de Fisiologia e Biofísica, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luciola S Barcelos
- Departamento de Fisiologia e Biofísica, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Diego C Dos Reis
- Departamento de Patologia Geral, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Geovanni D Cassali
- Departamento de Patologia Geral, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Maria E de Lima
- Programa de Pós-Graduação em Medicina-Biomedicina, Faculdade Santa Casa de Belo Horizonte, Belo Horizonte, Brazil
| | - Quênia J T de Castro
- Departamento de Farmácia, Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | - Andrea Grabe-Guimarães
- Departamento de Farmácia, Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | - Adilson D da Silva
- Departamento de Química, Universidade Federal de Juiz de Fora, Juiz de Fora, Brazil
| | | |
Collapse
|
9
|
Lawther AJ, Phillips AJK, Chung NC, Chang A, Ziegler AI, Debs S, Sloan EK, Walker AK. Disrupting circadian rhythms promotes cancer-induced inflammation in mice. Brain Behav Immun Health 2022; 21:100428. [PMID: 35199050 PMCID: PMC8851215 DOI: 10.1016/j.bbih.2022.100428] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 02/07/2022] [Accepted: 02/07/2022] [Indexed: 02/09/2023] Open
Abstract
Disruption of circadian rhythms occurs in rotating shift-work, jetlag, and in individuals with irregular sleep schedules. Circadian disruption is known to alter inflammatory responses and impair immune function. However, there is limited understanding of how circadian disruption modulates cancer-induced inflammation. Inflammation is a hallmark of cancer and is linked to worse prognosis and impaired brain function in cancer patients. Here, we investigated the effect of circadian disruption on cancer-induced inflammation in an orthotopic breast cancer model. Using a validated chronic jetlag protocol that advances the light-cycle by 8 h every 2 days to disrupt circadian rhythms, we found that circadian disruption alters cancer-induced inflammation in a tissue-specific manner, increasing inflammation in the body and brain while decreasing inflammation within the tumor tissue. Circadian disruption did not affect inflammation in mice without tumors, suggesting that the impact of circadian disruption may be particularly detrimental in the context of underlying inflammatory conditions, such as cancer. Importantly, circadian disruption did not affect tumor burden, suggesting that increased inflammation was not a result of increased cancer progression. Overall, these findings identify the importance of healthy circadian rhythms for limiting cancer-induced inflammation. Circadian disruption enhances cancer-induced inflammation in the body and brain. The profile of inflammatory cytokines altered by circadian disruption is tissue specific. Changes in inflammatory profiles by circadian disruption are not due to enhanced tumor burden.
Collapse
Affiliation(s)
- Adam J Lawther
- Laboratory of ImmunoPsychiatry, Neuroscience Research Australia, Randwick, New South Wales, 2031, Australia
| | - Andrew J K Phillips
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, VIC, 3800, Australia
| | - Ni-Chun Chung
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
| | - Aeson Chang
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
| | - Alexandra I Ziegler
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
| | - Sophie Debs
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, New South Wales, 2031, Australia
| | - Erica K Sloan
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia.,Division of Cancer Surgery, Peter MacCallum Cancer Centre, East Melbourne, VIC, 3002, Australia
| | - Adam K Walker
- Laboratory of ImmunoPsychiatry, Neuroscience Research Australia, Randwick, New South Wales, 2031, Australia.,Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia.,School of Psychiatry, University of New South Wales, Kensington, NSW, 2033, Australia
| |
Collapse
|
10
|
Walker WH, Kaper AL, Meléndez-Fernández OH, Bumgarner JR, Liu JA, Walton JC, DeVries AC, Nelson RJ. Time-restricted feeding alters the efficiency of mammary tumor growth. Chronobiol Int 2021; 39:535-546. [PMID: 34894935 DOI: 10.1080/07420528.2021.2011306] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Disruption of circadian rhythms has detrimental host consequences. Indeed, both clinical and foundational science demonstrate a clear relationship between disruption of circadian rhythms and cancer initiation and progression. Because timing of food intake can act as a zeitgeber (i.e., entrainment signal) for the circadian clock, and most individuals in the developed world have access to food at all times of the day in a "24/7" society, we sought to determine the effects of timing of food intake on mammary tumor growth. We hypothesized that restricting access to food to during the inactive phase would accelerate tumor growth. Adult female Balb/C mice received a unilateral orthotopic injection of murine mammary carcinoma 4T1 cells into the ninth inguinal mammary gland. Beginning on the day of tumor injection and continuing until the end of the experiment, mice were food restricted to their active phase (ZT12 (lights off)- ZT0 (lights on), inactive phase (ZT0 - ZT12), or had ad libitum access to food. Mice that were food restricted to their inactive phase displayed a significant increase in body mass on days 7 and 14 of tumor growth relative to active phase or ad libitum fed mice. Additionally, mice fed during their inactive phase demonstrated a 20% reduction in food consumption relative to mice fed during their active phase and a 17% reduction in food consumption relative to ab libitum fed mice. Tumor volume was not significantly different between groups. However, food restricting mice to their inactive phase increased mammary tumor growth efficiency (i.e., mg of tumor mass per gram of food intake) relative to mice fed during the active phase and approached significance (p = .06) relative to ad libitum fed mice. To determine a potential explanation for the increased tumor growth efficiency, we examined rhythms of activity and body temperature. Mice fed during the inactive phase displayed significantly disrupted daily activity and body temperature rhythms relative to both other feeding regimens. Together, these data demonstrate that improperly timed food intake can have detrimental consequences on mammary tumor growth likely via disrupted circadian rhythms.
Collapse
Affiliation(s)
- William H Walker
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia, USA
| | - Alexis L Kaper
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia, USA
| | | | - Jacob R Bumgarner
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia, USA
| | - Jennifer A Liu
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia, USA
| | - James C Walton
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia, USA
| | - A Courtney DeVries
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia, USA.,Department of Medicine, Division of Oncology/Hematology, West Virginia, USA.,WVU Cancer Institute, West Virginia University, Morgantown, West Virginia, USA
| | - Randy J Nelson
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia, USA
| |
Collapse
|
11
|
Walker WH, Sprowls SA, Bumgarner JR, Liu JA, Meléndez-Fernández OH, Walton JC, Lockman PR, DeVries AC, Nelson RJ. Circadian Influences on Chemotherapy Efficacy in a Mouse Model of Brain Metastases of Breast Cancer. Front Oncol 2021; 11:752331. [PMID: 34956876 PMCID: PMC8695439 DOI: 10.3389/fonc.2021.752331] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 11/19/2021] [Indexed: 11/30/2022] Open
Abstract
Chemotherapy is more effective in the treatment of peripheral tumors than brain metastases, likely reflecting the reduced ability of chemotherapy to cross the blood-brain barrier (BBB) and blood-tumor barrier at efficacious concentrations. Recent studies demonstrate circadian regulation of the BBB. Thus, we predicted that optimally timed chemotherapy would increase anti-tumor efficacy in a model of brain metastases of breast cancer (BMBC). First, we characterized novel daily alterations in BBB permeability to a commonly used chemotherapeutic, 14C-paclitaxel, within BMBC following injections given at four time points across the day. Peak and trough 14C-paclitaxel concentrations within BMBC occurred during the mid-dark phase and at the beginning of the light phase, respectively. Notably, chemotherapy injections during the dark phase increased cell death within BMBC and delayed onset of neurological symptoms relative to injections during the light phase. These data provide strong evidence for the beneficial effects of chrono-chemotherapy for the treatment of BMBC.
Collapse
Affiliation(s)
- William H. Walker
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| | - Samuel A. Sprowls
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, WV, United States
| | - Jacob R. Bumgarner
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| | - Jennifer A. Liu
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| | | | - James C. Walton
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| | - Paul R. Lockman
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, WV, United States
- WVU Cancer Institute, West Virginia University, Morgantown, WV, United States
| | - A. Courtney DeVries
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
- WVU Cancer Institute, West Virginia University, Morgantown, WV, United States
- Department of Medicine, Division of Oncology/Hematology, West Virginia University, Morgantown, WV, United States
| | - Randy J. Nelson
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| |
Collapse
|
12
|
Walker WH, Kvadas RM, May LE, Liu JA, Bumgarner JR, Walton JC, DeVries AC, Dauchy RT, Blask DE, Nelson RJ. Artificial Light at Night Reduces Anxiety-like Behavior in Female Mice with Exacerbated Mammary Tumor Growth. Cancers (Basel) 2021; 13:cancers13194860. [PMID: 34638343 PMCID: PMC8508227 DOI: 10.3390/cancers13194860] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/17/2021] [Accepted: 09/22/2021] [Indexed: 01/06/2023] Open
Abstract
Simple Summary Artificial light at night, initially assumed to be innocuous, is associated with an increased risk for developing mood disorders, sleep disturbances, and cancer. However, the influence of ALAN on affective behavior in tumor-bearing mice has not been investigated. Here, we demonstrate that ALAN reduces the latency to tumor onset and increases terminal tumor volume. Additionally, tumor-bearing mice housed in dark nights exhibit increased anxiety-like behavior which is prevented via housing in ALAN. Abstract Artificial light at night (ALAN) is a pervasive phenomenon. Although initially assumed to be innocuous, recent research has demonstrated its deleterious effects on physiology and behavior. Exposure to ALAN is associated with disruptions to sleep/wake cycles, development of mood disorders, metabolic disorders, and cancer. However, the influence of ALAN on affective behavior in tumor-bearing mice has not been investigated. We hypothesize that exposure to ALAN accelerates mammary tumor growth and predict that ALAN exacerbates negative affective behaviors in tumor-bearing mice. Adult (>8 weeks) female C3H mice received a unilateral orthotropic injection of FM3A mouse mammary carcinoma cells (1.0 × 105 in 100 μL) into the fourth inguinal mammary gland. Nineteen days after tumor inoculation, mice were tested for sucrose preference (anhedonia-like behavior). The following day, mice were subjected to an open field test (anxiety-like behavior), followed by forced swim testing (depressive-like behavior). Regardless of tumor status, mice housed in ALAN increased body mass through the first ten days. Tumor-bearing ALAN-housed mice demonstrated reduced latency to tumor onset (day 5) and increased terminal tumor volume (day 21). Exposure to ALAN reduced sucrose preference independent of tumor status. Additionally, tumor-bearing mice housed in dark nights demonstrated significantly increased anxiety-like behavior that was normalized via housing in ALAN. Together, these data reaffirm the negative effects of ALAN on tumorigenesis and demonstrate the potential anxiolytic effect of ALAN in the presence of mammary tumors.
Collapse
Affiliation(s)
- William H. Walker
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26505, USA; (R.M.K.); (L.E.M.); (J.A.L.); (J.R.B.); (J.C.W.); (A.C.D.); (R.J.N.)
- Correspondence:
| | - Raegan M. Kvadas
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26505, USA; (R.M.K.); (L.E.M.); (J.A.L.); (J.R.B.); (J.C.W.); (A.C.D.); (R.J.N.)
| | - Laura E. May
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26505, USA; (R.M.K.); (L.E.M.); (J.A.L.); (J.R.B.); (J.C.W.); (A.C.D.); (R.J.N.)
| | - Jennifer A. Liu
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26505, USA; (R.M.K.); (L.E.M.); (J.A.L.); (J.R.B.); (J.C.W.); (A.C.D.); (R.J.N.)
| | - Jacob R. Bumgarner
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26505, USA; (R.M.K.); (L.E.M.); (J.A.L.); (J.R.B.); (J.C.W.); (A.C.D.); (R.J.N.)
| | - James C. Walton
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26505, USA; (R.M.K.); (L.E.M.); (J.A.L.); (J.R.B.); (J.C.W.); (A.C.D.); (R.J.N.)
| | - A. Courtney DeVries
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26505, USA; (R.M.K.); (L.E.M.); (J.A.L.); (J.R.B.); (J.C.W.); (A.C.D.); (R.J.N.)
- Department of Medicine, Division of Oncology/Hematology, West Virginia University, Morgantown, WV 26505, USA
- Cancer Institute, West Virginia University, Morgantown, WV 26505, USA
| | - Robert T. Dauchy
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA; (R.T.D.); (D.E.B.)
| | - David E. Blask
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA; (R.T.D.); (D.E.B.)
| | - Randy J. Nelson
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26505, USA; (R.M.K.); (L.E.M.); (J.A.L.); (J.R.B.); (J.C.W.); (A.C.D.); (R.J.N.)
| |
Collapse
|
13
|
Abdel-Salam MAL, Pinto B, Cassali G, Bueno L, Pêgas G, Oliveira F, Silva I, Klein A, de Souza-Fagundes EM, de Lima ME, Carvalho-Tavares J. LyeTx I-b Peptide Attenuates Tumor Burden and Metastasis in a Mouse 4T1 Breast Cancer Model. Antibiotics (Basel) 2021; 10:1136. [PMID: 34572719 PMCID: PMC8466574 DOI: 10.3390/antibiotics10091136] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/11/2021] [Accepted: 09/16/2021] [Indexed: 12/20/2022] Open
Abstract
Cationic anticancer peptides have exhibited potent anti-proliferative and anti-inflammatory effects in neoplastic illness conditions. LyeTx I-b is a synthetic peptide derived from Lycosa erythrognatha spider venom that previously showed antibiotic activity in vitro and in vivo. This study focused on the effects of LyeTxI-b on a 4T1 mouse mammary carcinoma model. Mice with a palpable tumor in the left flank were subcutaneously or intratumorally injected with LyeTx I-b (5 mg/kg), which significantly decreased the tumor volume and metastatic nodules. Histological analyses showed a large necrotic area in treated primary tumors compared to the control. LyeTxI-b reduced tumor growth and lung metastasis in the 4T1 mouse mammary carcinoma model with no signs of toxicity in healthy or cancerous mice. The mechanism of action of LyeTx I-b on the 4T1 mouse mammary carcinoma model was evaluated in vitro and is associated with induction of apoptosis and cell proliferation inhibition. Furthermore, LyeTx I-b seems to be an efficient regulator of the 4T1 tumor microenvironment by modulating several cytokines, such as TGF-β, TNF-α, IL-1β, IL-6, and IL-10, in primary tumor and lung, spleen, and brain. LyeTx I-b also plays a role in leukocytes rolling and adhesion into spinal cord microcirculation and in the number of circulating leukocytes. These data suggest a potent antineoplastic efficacy ofLyeTx I-b.
Collapse
Affiliation(s)
- Mostafa A. L. Abdel-Salam
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia e Farmacologia, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (M.A.L.A.-S.); (B.P.); (E.M.d.S.-F.)
| | - Bárbara Pinto
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia e Farmacologia, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (M.A.L.A.-S.); (B.P.); (E.M.d.S.-F.)
| | - Geovanni Cassali
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (G.C.); (G.P.); (F.O.)
| | - Lilian Bueno
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil;
| | - Gabriela Pêgas
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (G.C.); (G.P.); (F.O.)
| | - Fabrício Oliveira
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (G.C.); (G.P.); (F.O.)
| | - Irismara Silva
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia e Farmacologia, Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (I.S.); (A.K.)
| | - André Klein
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia e Farmacologia, Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (I.S.); (A.K.)
| | - Elaine Maria de Souza-Fagundes
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia e Farmacologia, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (M.A.L.A.-S.); (B.P.); (E.M.d.S.-F.)
| | - Maria Elena de Lima
- Programa de Pós-Graduação em Medicina-Biomedicina, Faculdade Santa Casa de Belo Horizonte, Belo Horizonte 30110-005, Brazil
| | - Juliana Carvalho-Tavares
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia e Farmacologia, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (M.A.L.A.-S.); (B.P.); (E.M.d.S.-F.)
| |
Collapse
|
14
|
Mampay M, Flint MS, Sheridan GK. Tumour brain: Pretreatment cognitive and affective disorders caused by peripheral cancers. Br J Pharmacol 2021; 178:3977-3996. [PMID: 34029379 DOI: 10.1111/bph.15571] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 04/12/2021] [Accepted: 05/05/2021] [Indexed: 12/20/2022] Open
Abstract
People that develop extracranial cancers often display co-morbid neurological disorders, such as anxiety, depression and cognitive impairment, even before commencement of chemotherapy. This suggests bidirectional crosstalk between non-CNS tumours and the brain, which can regulate peripheral tumour growth. However, the reciprocal neurological effects of tumour progression on brain homeostasis are not well understood. Here, we review brain regions involved in regulating peripheral tumour development and how they, in turn, are adversely affected by advancing tumour burden. Tumour-induced activation of the immune system, blood-brain barrier breakdown and chronic neuroinflammation can lead to circadian rhythm dysfunction, sleep disturbances, aberrant glucocorticoid production, decreased hippocampal neurogenesis and dysregulation of neural network activity, resulting in depression and memory impairments. Given that cancer-related cognitive impairment diminishes patient quality of life, reduces adherence to chemotherapy and worsens cancer prognosis, it is essential that more research is focused at understanding how peripheral tumours affect brain homeostasis.
Collapse
Affiliation(s)
- Myrthe Mampay
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, UK
| | - Melanie S Flint
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, UK
| | - Graham K Sheridan
- School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| |
Collapse
|
15
|
Carter T, Qi G, Wang W, Nguyen A, Cheng N, Ju YM, Lee SJ, Yoo JJ, Atala A, Sun XS. Self-Assembling Peptide Solution Accelerates Hemostasis. Adv Wound Care (New Rochelle) 2021; 10:191-203. [PMID: 32716728 DOI: 10.1089/wound.2019.1109] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Objective: One of the leading causes of death following traumatic injury is exsanguination. Biological material-based hemostatic agents such as fibrin, thrombin, and albumin have a high risk for causing infection. Synthetic peptide-based hemostatic agents offer an attractive alternative. The objective of this study is to explore the potential of h9e peptide as an effective hemostatic agent in both in vitro and in vivo models. Approach: In vitro blood coagulation kinetics in the presence of h9e peptide was determined as a function of gelation time using a dynamic rheometer. In vivo hemostatic effects were studied using the Wistar rat model. Results were compared to those of the commercial hemostatic product Celox™, a chitosan-based product. Adhesion of h9e peptide was evaluated using the platelet adhesion test. Biocompatibility of h9e peptide was studied in vivo using a mouse model. Results: After h9e peptide solution was mixed with blood, gelation started immediately, increased rapidly with time, and reached more than 100 Pa within 3 s. Blood coagulation strength increased as h9e peptide wt% concentration increased. In the rat model, h9e peptide solution at 5% weight concentration significantly reduced both bleeding time and blood loss, outperforming Celox. Preliminary pathological studies indicate that h9e peptide solution is biocompatible and did not have negative effects when injected subcutaneously in a mouse model. Innovation: For the first time, h9e peptide was found to have highly efficient hemostatic effects by forming nanoweb-like structures, which act as a preliminary thrombus and a surface to arrest bleeding 82% faster compared to the commercial hemostatic agent Celox. Conclusion: This study demonstrates that h9e peptide is a promising hemostatic biomaterial, not only because of its greater hemostatic effect than commercial product Celox but also because of its excellent biocompatibility based on the in vivo mouse model study.
Collapse
Affiliation(s)
- Tiffany Carter
- Bio-Materials and Technology Lab, Grain Science and Industry, Kansas State University, Manhattan, Kansas, USA
- Department of Agriculture, Austin Peay State University, Clarksville, Tennessee, USA
| | - Guangyan Qi
- Bio-Materials and Technology Lab, Grain Science and Industry, Kansas State University, Manhattan, Kansas, USA
| | - Weiqun Wang
- Human Nutrition, Kansas State University, Manhattan, Kansas, USA
| | - Annelise Nguyen
- Diagnostic Medicine and Pathology, Kansas State University, Manhattan, Kansas, USA
| | - Nikki Cheng
- Pathology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Young Min Ju
- Wake Forest Institute of Regenerative Medicine, School of Medicine, Wake Forest University, Winston-Salem, North Carolina, USA
| | - Sang Jin Lee
- Wake Forest Institute of Regenerative Medicine, School of Medicine, Wake Forest University, Winston-Salem, North Carolina, USA
| | - James J. Yoo
- Wake Forest Institute of Regenerative Medicine, School of Medicine, Wake Forest University, Winston-Salem, North Carolina, USA
| | - Anthony Atala
- Wake Forest Institute of Regenerative Medicine, School of Medicine, Wake Forest University, Winston-Salem, North Carolina, USA
| | - Xiuzhi Susan Sun
- Bio-Materials and Technology Lab, Grain Science and Industry, Kansas State University, Manhattan, Kansas, USA
- Wake Forest Institute of Regenerative Medicine, School of Medicine, Wake Forest University, Winston-Salem, North Carolina, USA
- Biological and Agricultural Engineering, Kansas State University, Manhattan, Kansas, USA
| |
Collapse
|
16
|
Castro F, Martins C, Silveira MJ, Moura RP, Pereira CL, Sarmento B. Advances on erythrocyte-mimicking nanovehicles to overcome barriers in biological microenvironments. Adv Drug Deliv Rev 2021; 170:312-339. [PMID: 32946921 DOI: 10.1016/j.addr.2020.09.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/29/2020] [Accepted: 09/05/2020] [Indexed: 12/14/2022]
Abstract
Although nanocarriers offer many advantages as drug delivery systems, their poor stability in circulation, premature drug release and nonspecific uptake in non-target organs have prompted biomimetic approaches using natural cell membranes to camouflage nanovehicles. Among them, erythrocytes, representing the most abundant blood circulating cells, have been extensively investigated for biomimetic coating on artificial nanocarriers due to their upgraded biocompatibility, biodegradability, non-immunogenicity and long-term blood circulation. Due to the cell surface mimetic properties combined with customized core material, erythrocyte-mimicking nanovehicles (EM-NVs) have a wide variety of applications, including drug delivery, imaging, phototherapy, immunomodulation, sensing and detection, that foresee a huge potential for therapeutic and diagnostic applications in several diseases. In this review, we summarize the recent advances in the biomedical applications of EM-NVs in cancer, infection, heart-, autoimmune- and CNS-related disorders and discuss the major challenges and opportunities in this research area.
Collapse
Affiliation(s)
- Flávia Castro
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Cláudia Martins
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Maria José Silveira
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Rui Pedro Moura
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal; CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Catarina Leite Pereira
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Bruno Sarmento
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal.
| |
Collapse
|
17
|
Shakibapour M, Shojaie B, Yousofi Darani H. Immunization with Hydatid Cyst Wall Antigens Can Inhibit Breast Cancer through Changes in Serum Levels of Th1/Th2 Cytokines. Int J Prev Med 2020; 11:189. [PMID: 33815713 PMCID: PMC8000162 DOI: 10.4103/ijpvm.ijpvm_311_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 10/31/2019] [Indexed: 11/04/2022] Open
Abstract
Background Hydatid cysts are the larval stage of Echinococcus granulosus, which lead to humoral and cellular immune responses in hosts. Such immune responses play a key role in the inhibition of tumor growth and cancers. To test this hypothesis, it was attempted not only to examine the changes in serum level of some Th1 and Th2 cytokines but also to find relationships between the cytokines and cancer in 4T1 breast cancer-bearing mice immunized with hydatid cyst wall (HCW) antigens. Methods Six to eight-week-old Balb/c female mice were immunized with alum, PBS and HCW antigens, including crude extract of HCW (laminated layer) 28 and 27 kDa protein bands (upper and lower bands) and then challenged with 4T1 breast cancer cells. The amounts of IL2, TNF-α, IFN-γ (Th1 cytokines), and IL4 (Th2 cytokine) were estimated using ELISA. Correlations between these cytokines and cancer parameters (tumor growth, metastasis, and survival) were determined by Pearson's correlation coefficients. Results Overall, HCW antigens increased the amounts of IL2, TNF-α, IFN-γ, and IL4. Pearson's correlation coefficients indicated reverse relationships between changes in amounts of these cytokines and tumor growth/metastasis. However, except for IL-4, all cytokines had a direct relationship with mouse survival. Conclusions The results of this study indicated that the inhibition of breast tumor growth and metastasis and improvement of survival in 4T1 mice immunized with HCW antigens, especially laminated layer and 27 kDa protein band can occur through a rise in the levels of cytokines.
Collapse
Affiliation(s)
- Mahshid Shakibapour
- Department of Medical Parasitology and Mycology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Behrokh Shojaie
- Department of Medical Parasitology and Mycology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.,Department of Biology, Faculty of Science, University of Isfahan, Isfahan, Iran
| | - Hossein Yousofi Darani
- Department of Medical Parasitology and Mycology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
18
|
Walker WH, Meléndez-Fernández OH, Pascoe JL, Zhang N, DeVries AC. Social enrichment attenuates chemotherapy induced pro-inflammatory cytokine production and affective behavior via oxytocin signaling. Brain Behav Immun 2020; 89:451-464. [PMID: 32735935 PMCID: PMC7572590 DOI: 10.1016/j.bbi.2020.07.032] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 12/24/2022] Open
Abstract
Breast cancer survivors receiving chemotherapy often report increased anxiety and depression. However, the mechanism underlying chemotherapy-induced changes in affect remains unknown. We hypothesized that chemotherapy increases cytokine production, in turn altering exploratory and depressive-like behavior. To test this hypothesis, female Balb/C mice received two injections, separated by two weeks, of vehicle (0.9% saline) or a chemotherapeutic cocktail [9 mg/kg doxorubicin (A) and 90 mg/kg cyclophosphamide (C)]. Peripheral and central cytokine concentrations were increased one and seven days, respectively, after AC. Because of the beneficial effects of social enrichment on several diseases with inflammatory components, we examined whether social enrichment could attenuate the increase in peripheral and central cytokine production following chemotherapy administration. Socially isolated mice receiving AC therapy demonstrated increased depressive-like and exploratory behaviors with a concurrent increase in hippocampal IL-6. Whereas, group housing attenuated AC-induced IL-6 and depressive-like behavior. Next, we sought to determine whether central oxytocin may contribute to the protective effects of social housing after AC administration. Intracerebroventricular administration of oxytocin to socially isolated mice recapitulated the protective effects of social enrichment; specifically, oxytocin ameliorated the AC-induced effects on IL-6 and depressive-like behavior. Furthermore, administration of an oxytocin antagonist to group housed mice recapitulated the responses of socially isolated mice; specifically, AC increased depressive-like behavior and central IL-6. These data suggest a possible neuroprotective role for oxytocin following chemotherapy, via modulation of IL-6. This study adds to the growing literature detailing the negative behavioral effects of chemotherapy and provides further evidence that social enrichment may be beneficial to health.
Collapse
Affiliation(s)
- William H. Walker
- Department of Neuroscience, West Virginia University, Morgantown, WV, 26506 USA,Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, 26506 USA,Corresponding Author:, 108 Biomedical Road, BMRC Room 370, Morgantown, WV 26506 USA
| | - O. Hecmarie Meléndez-Fernández
- Department of Neuroscience, West Virginia University, Morgantown, WV, 26506 USA,Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, 26506 USA
| | - Jordan L. Pascoe
- Department of Neuroscience, West Virginia University, Morgantown, WV, 26506 USA,Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, 26506 USA
| | - Ning Zhang
- Department of Neuroscience, West Virginia University, Morgantown, WV, 26506 USA,Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, 26506 USA
| | - A. Courtney DeVries
- Department of Neuroscience, West Virginia University, Morgantown, WV, 26506 USA,Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, 26506 USA,West Virginia University Cancer Institute, West Virginia University, Morgantown, WV, 26506 USA,Department of Medicine, West Virginia University, Morgantown, WV, 26506 USA
| |
Collapse
|
19
|
Castro F, Pinto ML, Pereira CL, Serre K, Barbosa MA, Vermaelen K, Gärtner F, Gonçalves RM, De Wever O, Oliveira MJ. Chitosan/γ-PGA nanoparticles-based immunotherapy as adjuvant to radiotherapy in breast cancer. Biomaterials 2020; 257:120218. [DOI: 10.1016/j.biomaterials.2020.120218] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/05/2020] [Accepted: 06/22/2020] [Indexed: 02/07/2023]
|
20
|
Li J, Liu M, Gao J, Jiang Y, Wu L, Cheong YK, Ren G, Yang Z. AVNP2 protects against cognitive impairments induced by C6 glioma by suppressing tumour associated inflammation in rats. Brain Behav Immun 2020; 87:645-659. [PMID: 32097763 PMCID: PMC7126810 DOI: 10.1016/j.bbi.2020.02.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 01/24/2020] [Accepted: 02/20/2020] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma is a kind of malignant tumour and originates from the central nervous system. In the last century, some researchers and clinician have noticed that the psychosocial and neurocognitive functioning of patients with malignant gliomas can be impaired. Many clinical studies have demonstrated that part of patients, adults or children, diagnosed with glioblastoma will suffer from cognitive deficiency during their clinical course, especially in long-term survivors. Many nanoparticles (NPs) can inhibit the biological functions of tumours by modulating tumour-associated inflammation, which provokes angiogenesis and tumour growth. As one of the best antiviral nanoparticles (AVNPs), AVNP2 is the 2nd generation of AVNP2 that have been conjugated to graphite-graphene for improving physiochemical performance and reducing toxicity. AVNP2 inactivates viruses, such as the H1N1 and H5N1influenza viruses and even the SARS coronavirus, while it inhibits bacteria, such as MRSA and E. coli. As antimicrobials, nanoparticles are considered to be one of the vectors for the administration of therapeutic compounds. Yet, little is known about their potential functionalities and toxicities to the neurotoxic effects of cancer. Herein, we explored the functionality of AVNP2 on inhibiting C6 in glioma-bearing rats. The novel object-recognition test and open-field test showed that AVNP2 significantly improved the neuro-behaviour affected by C6 glioma. AVNP2 also alleviated the decline of long-term potentiation (LTP) and the decreased density of dendritic spines in the CA1 region induced by C6. Western blot assay and immunofluorescence staining showed that the expressions of synaptic-related proteins (PSD-95 and SYP) were increased, and these findings were in accordance with the results mentioned above. It revealed that the sizes of tumours in C6 glioma-bearing rats were smaller after treatment with AVNP2. The decreased expression of inflammatory factors (IL-1β, IL-6 and TNF-α) by Western blotting assay and ELISA, angiogenesis protein (VEGF) by Western blotting assay and other related proteins (BDNF, NF-ĸB, iNOS and COX-2) by Western blotting assay in peri-tumour tissue indicated that AVNP2 could control tumour-associated inflammation, thus efficiently ameliorating the local inflammatory condition and, to some extent, inhibiting angiogenesis in C6-bearing rats. In conclusion, our results suggested that AVNP2 could have an effect on the peri-tumor environment, obviously restraining the growth progress of gliomas, and eventually improving cognitive levels in C6-bearing rats.
Collapse
Affiliation(s)
- Junyang Li
- Medical School, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, Nankai University, Tianjin 300071, China
| | - Meicen Liu
- Medical School, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, Nankai University, Tianjin 300071, China
| | - Jin Gao
- Medical School, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, Nankai University, Tianjin 300071, China
| | - Yu Jiang
- Medical School, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, Nankai University, Tianjin 300071, China
| | - Limin Wu
- Institute of Laser and Optoelectronics, School of Precision Instruments and Optoelectronics Engineering, Tianjin University, Tianjin 300072, China
| | - Yuen-Ki Cheong
- Science and Technology Research Institute, University of Hertfordshire, Hatfield, Herts AL10 9AB, UK
| | - Guogang Ren
- Science and Technology Research Institute, University of Hertfordshire, Hatfield, Herts AL10 9AB, UK
| | - Zhuo Yang
- Medical School, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, Nankai University, Tianjin 300071, China.
| |
Collapse
|
21
|
Walker WH, Borniger JC, Gaudier-Diaz MM, Hecmarie Meléndez-Fernández O, Pascoe JL, Courtney DeVries A, Nelson RJ. Acute exposure to low-level light at night is sufficient to induce neurological changes and depressive-like behavior. Mol Psychiatry 2020; 25:1080-1093. [PMID: 31138889 PMCID: PMC6881534 DOI: 10.1038/s41380-019-0430-4] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 04/12/2019] [Accepted: 04/29/2019] [Indexed: 12/20/2022]
Abstract
The advent and wide-spread adoption of electric lighting over the past century has profoundly affected the circadian organization of physiology and behavior for many individuals in industrialized nations; electric lighting in homes, work environments, and public areas have extended daytime activities into the evening, thus, increasing night-time exposure to light. Although initially assumed to be innocuous, chronic exposure to light at night (LAN) is now associated with increased incidence of cancer, metabolic disorders, and affective problems in humans. However, little is known about potential acute effects of LAN. To determine whether acute exposure to low-level LAN alters brain function, adult male, and female mice were housed in either light days and dark nights (LD; 14 h of 150 lux:10 h of 0 lux) or light days and low level light at night (LAN; 14 h of 150 lux:10 h of 5 lux). Mice exposed to LAN on three consecutive nights increased depressive-like responses compared to mice housed in dark nights. In addition, female mice exposed to LAN increased central tendency in the open field. LAN was associated with reduced hippocampal vascular endothelial growth factor-A (VEGF-A) in both male and female mice, as well as increased VEGFR1 and interleukin-1β mRNA expression in females, and reduced brain derived neurotrophic factor mRNA in males. Further, LAN significantly altered circadian rhythms (activity and temperature) and circadian gene expression in female and male mice, respectively. Altogether, this study demonstrates that acute exposure to LAN alters brain physiology and can be detrimental to well-being in otherwise healthy individuals.
Collapse
Affiliation(s)
- William H Walker
- Department of Medicine, Division of Oncology/Hematology, West Virginia University, Morgantown, WV, 26506, USA.
- Department of Neuroscience, West Virginia University, Morgantown, WV, 26506, USA.
- Department of Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, 26506, USA.
- Department of Neuroscience Graduate Program, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA.
| | - Jeremy C Borniger
- Department of Neuroscience Graduate Program, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Monica M Gaudier-Diaz
- Department of Neuroscience Graduate Program, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Department of Psychology and Neuroscience, University of North Carolina Chapel Hill, Chapel Hill, NC, 27514, USA
| | - O Hecmarie Meléndez-Fernández
- Department of Neuroscience, West Virginia University, Morgantown, WV, 26506, USA
- Department of Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, 26506, USA
| | - Jordan L Pascoe
- Department of Neuroscience, West Virginia University, Morgantown, WV, 26506, USA
- Department of Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, 26506, USA
| | - A Courtney DeVries
- Department of Medicine, Division of Oncology/Hematology, West Virginia University, Morgantown, WV, 26506, USA
- Department of Neuroscience, West Virginia University, Morgantown, WV, 26506, USA
- Department of Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, 26506, USA
| | - Randy J Nelson
- Department of Neuroscience, West Virginia University, Morgantown, WV, 26506, USA
- Department of Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, 26506, USA
| |
Collapse
|
22
|
Casaril AM, Domingues M, Bampi SR, Lourenço DDA, Smaniotto TÂ, Segatto N, Vieira B, Seixas FK, Collares T, Lenardão EJ, Savegnago L. The antioxidant and immunomodulatory compound 3-[(4-chlorophenyl)selanyl]-1-methyl-1H-indole attenuates depression-like behavior and cognitive impairment developed in a mouse model of breast tumor. Brain Behav Immun 2020; 84:229-241. [PMID: 31837417 DOI: 10.1016/j.bbi.2019.12.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 11/20/2019] [Accepted: 12/09/2019] [Indexed: 12/15/2022] Open
Abstract
Psychiatric alterations are often found in patients with breast cancer even before the initiation of adjuvant therapy, resulting in a poor quality of life. It has become accepted that neuroinflammation and oxidative stress are involved in the pathophysiology of depression and cognitive impairment. Herein, we tested the hypothesis that treatment with the antioxidant and immunomodulatory selenium-containing compound 3-[(4-chlorophenyl)selanyl]-1-methyl-1H-indole (CMI)could attenuate behavioral and neurochemical alterations in a mammary (4T1) tumor model. Female BALB/c mice were subcutaneously inoculated with 4T1 cancer cells (1 × 105 cells/mice) or PBS. From days 14 to 20, mice received daily gavage with canola oil or CMI. On day 21, mice were submitted to behavioral tests followed by euthanasia. We found that CMI did not alter tumor growth, body weight, and body temperature in tumor-bearing mice. Importantly, treatment with CMI abrogated tumor-induced depression-like behavior and cognitive impairment. By the time CMI improved the behavioral alterations, it had reduced tumor-induced neuroinflammation (altered expression of NFκB, IL-1β, TNF-α, IL-10, IDO, and COX-2) and oxidative stress (altered expression of iNOS and Nrf2, and levels of reactive species, nitric oxide, lipid peroxidation, and superoxide dismutase activity) in the prefrontal cortices and hippocampi of mice. A molecular docking approach suggested the ability of CMI to inhibit the activity of iNOS and COX-2. Together, our results indicate that CMI treatment may attenuate depression and cognitive impairment in 4T1 tumor-bearing mice, and be a groundbreaking strategy for the treatment of cancer-related psychiatric symptoms to improve the quality of life of cancer patients.
Collapse
Affiliation(s)
- Angela Maria Casaril
- Technological Development Center, Division of Biotechnology, Neurobiotechology Research Group, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Micaela Domingues
- Technological Development Center, Division of Biotechnology, Neurobiotechology Research Group, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Suely Ribeiro Bampi
- Technological Development Center, Division of Biotechnology, Neurobiotechology Research Group, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Darling de Andrade Lourenço
- Technological Development Center, Division of Biotechnology, Neurobiotechology Research Group, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Thiago Ângelo Smaniotto
- Technological Development Center, Division of Biotechnology, Neurobiotechology Research Group, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Natália Segatto
- Technological Development Center, Division of Biotechnology, Molecular and Cellular Oncology Research Group and Functional Genomics Laboratory, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Beatriz Vieira
- Center of Chemical, Pharmaceutical and Food Sciences, Laboratory of Clean Organic Synthesis, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Fabiana K Seixas
- Technological Development Center, Division of Biotechnology, Molecular and Cellular Oncology Research Group and Functional Genomics Laboratory, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Tiago Collares
- Technological Development Center, Division of Biotechnology, Molecular and Cellular Oncology Research Group and Functional Genomics Laboratory, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Eder João Lenardão
- Center of Chemical, Pharmaceutical and Food Sciences, Laboratory of Clean Organic Synthesis, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Lucielli Savegnago
- Technological Development Center, Division of Biotechnology, Neurobiotechology Research Group, Federal University of Pelotas, Pelotas, RS, Brazil.
| |
Collapse
|
23
|
Walker WH, Bumgarner JR, Nelson RJ, Courtney DeVries A. Transcardial perfusion is not required to accurately measure cytokines within the brain. J Neurosci Methods 2020; 334:108601. [PMID: 31981570 PMCID: PMC7374017 DOI: 10.1016/j.jneumeth.2020.108601] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/21/2020] [Accepted: 01/21/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Cytokines are key signaling molecules within the immune system that regulate a host's response to pathogens and neuronal damage. Aberrant cytokine signaling has been implicated in many neurological diseases. Therefore, accurately measuring cytokine concentrations within the brain is crucial. NEW METHOD This study demonstrates that removing blood within brain vasculature via saline perfusion does not alter brain parenchymal cytokine protein concentrations or mRNA expression. RESULTS Hippocampal protein and mRNA data demonstrate that brain parenchymal cytokine concentrations do not significantly differ based on the method of euthanasia (i.e., perfusion or no perfusion). These results are consistent within naive and immune challenged mice. COMPARISON WITH EXISTING METHOD Due to the potential of cytokine contamination from circulating blood, it is believed that transcardial perfusion is required for accurate measurement of cytokine concentrations and gene expression within the brain. However, our data indicate that cytokine concentrations are unaffected by not perfusing mice with saline prior to tissue collection. CONCLUSIONS Brain cytokine concentrations are unaffected by perfusing with saline prior to tissue collection; this holds true regardless of immune status (homeostatic or immune challenged), suggesting that this time-consuming step may be unnecessary.
Collapse
Affiliation(s)
- William H Walker
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, 108 Biomedical Road, Morgantown, WV 26505 USA.
| | - Jacob R Bumgarner
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, 108 Biomedical Road, Morgantown, WV 26505 USA
| | - Randy J Nelson
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, 108 Biomedical Road, Morgantown, WV 26505 USA
| | - A Courtney DeVries
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, 108 Biomedical Road, Morgantown, WV 26505 USA; Department of Medicine, West Virginia University, 108 Biomedical Road, Morgantown, WV 26505 USA
| |
Collapse
|
24
|
Santos JC, Bever SR, Sullivan KA, Pyter LM. Cancer and cancer survival modulates brain and behavior in a time-of-day-dependent manner in mice. Sci Rep 2019; 9:6497. [PMID: 31019214 PMCID: PMC6482139 DOI: 10.1038/s41598-019-42880-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 04/10/2019] [Indexed: 01/01/2023] Open
Abstract
Improvements in breast cancer therapy/diagnosis have substantially increased the cancer survivor population, although many survivors report persistent mental health issues including fatigue, mood and anxiety disorders, and cognitive impairments. These behavioral symptoms impair quality-of-life and are often associated with increased inflammation. Nocturnal rodent models of cancer are critical to the identification of the neurobiological mechanisms underlying these behavioral changes. Although both behavior and immunity display distinct diurnal patterns, most rodent research in this field is performed during the rodents’ inactive (light) period, which could potentially undermine the conclusions and clinical relevance. Therefore, here we tested the extent to which mammary tumors or tumor resection (“survivors”) in mice affects behavior and neuroinflammation in a nyctohemeral (day versus night)-dependent manner. Indeed, only the dark (active) phase unmasked fatigue-like behavior and altered novel object investigation for both tumor-bearing and -resected mice relative to surgical controls. Several inflammatory markers were expressed in a time-of-day-dependent manner (lower in the dark phase) in the blood and brains of surgical control mice, whereas this temporal pattern was absent (IL-1β, CXCL1, Myd88, Cd4) or reversed (C3) in the respective tissues of tumor-bearing and -resected mice. Taken together, these data indicate that the time of day of assessment significantly modulates various persistent and transient tumor-induced behavioral and immune changes.
Collapse
Affiliation(s)
- Jessica C Santos
- Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, OH, USA.,Postgraduate Program in Basic and Applied Immunology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Savannah R Bever
- Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, OH, USA.,Department of Psychiatry and Behavioral Health, Ohio State University, Columbus, OH, USA
| | - Kyle A Sullivan
- Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, OH, USA.,Department of Neuroscience, Ohio State University, Columbus, OH, USA
| | - Leah M Pyter
- Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, OH, USA. .,Department of Psychiatry and Behavioral Health, Ohio State University, Columbus, OH, USA. .,Department of Neuroscience, Ohio State University, Columbus, OH, USA.
| |
Collapse
|
25
|
Santos JC, Bever SR, Pereira-da-Silva G, Pyter LM. Tumor resection ameliorates tumor-induced suppression of neuroinflammatory and behavioral responses to an immune challenge in a cancer survivor model. Sci Rep 2019; 9:752. [PMID: 30679700 PMCID: PMC6345941 DOI: 10.1038/s41598-018-37334-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 11/30/2018] [Indexed: 12/22/2022] Open
Abstract
Breast cancer survivors display altered inflammatory responses to immune challenges relative to cancer-naive controls likely due to previous cancer treatments, stress associated with cancer, and/or tumor physiology. Proper inflammatory responses are necessary for adaptive sickness behaviors (e.g., fatigue, anorexia, and fever) and neuroinflammatory pathways are also implicated in mental health disturbances (e.g., cognitive impairment, depression) suffered by cancer patients and survivors. Rodent cancer models indicate that tumors are sufficient to exacerbate neuroinflammatory responses after an immune challenge, however primary tumors are not usually present in cancer survivors, and the behavioral consequences of these brain changes remain understudied. Therefore, we tested the extent to which mammary tumor resection attenuates tumor-induced neuroinflammation and sickness behavior following an immune challenge (i.p. lipopolysaccharide [LPS] injection) in mice. Tnf-α, Il-1β, and Il-6 mRNA decreased in multiple brain regions of LPS-treated tumor-bearing mice relative to LPS-treated controls; tumor resection attenuated these effects in some cases (but not Tnf-α). Tumors also attenuated sickness behaviors (hypothermia and lethargy) compared to LPS-treated controls. Tumor resection reversed these behavioral consequences, although basal body temperature remained elevated, comparable to tumor-bearing mice. Thus, tumors significantly modulate neuroinflammatory pathways with functional consequences and tumor resection mitigates most, but not all, of these changes.
Collapse
Affiliation(s)
- Jessica C Santos
- Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, OH, USA.,Postgraduate Program in Basic and Applied Immunology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Savannah R Bever
- Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, OH, USA.,Department of Psychiatry and Behavioral Health, Ohio State University, Columbus, OH, USA
| | - Gabriela Pereira-da-Silva
- Postgraduate Program in Basic and Applied Immunology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, SP, Brazil.,Department of Maternal-Infant Nursing and Public Health, Ribeirão Preto College of Nursing, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Leah M Pyter
- Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, OH, USA. .,Department of Psychiatry and Behavioral Health, Ohio State University, Columbus, OH, USA. .,Department of Neuroscience, Ohio State University, Columbus, OH, USA. .,Arthur G. James Comprehensive Cancer Center and Solove Research institute, Ohio State University, Columbus, OH, USA.
| |
Collapse
|
26
|
Emmer KM, Walker WH, Zhang N, DeVries AC. Mammary tumor and mastectomy synergistically promote neuroinflammation in a breast cancer survivor model. Brain Res 2018; 1707:133-140. [PMID: 30496733 DOI: 10.1016/j.brainres.2018.11.037] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 11/21/2018] [Accepted: 11/25/2018] [Indexed: 12/23/2022]
Abstract
Understanding why breast cancer survivors are at an increased risk for cognitive and affective disorders is essential for developing targeted treatment plans and improving quality of life. Microglia priming results in chronic neuroinflammation and can contribute to neuronal degeneration and dysfunction, thereby offering a potential mechanism for altered brain function that persists after tumor removal. This study examined whether mammary tumors alter microglia and augment the inflammatory profile and behavior of mice. To test this, non-metastatic mammary tumor cells (67NR) were injected orthotopically into the mammary glands of BALB/c mice, allowed to grow for 16 days, and then the tumors were removed via mastectomy. Following a 14-day surgical recovery, the mice were challenged with lipopolysaccharide (LPS), and then central and peripheral inflammation, anxiety, and depressive-like behavior were evaluated. Here we show that major central and peripheral inflammatory markers were not altered by tumor growth nor mastectomy surgery alone. However, hippocampal mRNA expression of major proinflammatory cytokines IL-1β and TNFα was increased in tumor removal animals, persisting past surgical recovery. Nonetheless, the immune and behavioral responses following LPS administration were comparable among groups. In sum, these data demonstrate that the combination of tumor and mastectomy promotes neuroinflammation; however, immune challenge did not elucidate this inflammation as maladaptive for the host.
Collapse
Affiliation(s)
- Kathryn M Emmer
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, United States; Department of Veterinary Preventative Medicine, The Ohio State University, Columbus, OH, United States.
| | - William H Walker
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, United States; Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States; Department of Medicine, West Virginia University, Morgantown, WV, United States
| | - Ning Zhang
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, United States; Department of Medicine, West Virginia University, Morgantown, WV, United States
| | - A Courtney DeVries
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States; Department of Medicine, West Virginia University, Morgantown, WV, United States
| |
Collapse
|
27
|
Santos JC, Pyter LM. Neuroimmunology of Behavioral Comorbidities Associated With Cancer and Cancer Treatments. Front Immunol 2018; 9:1195. [PMID: 29930550 PMCID: PMC6001368 DOI: 10.3389/fimmu.2018.01195] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 05/14/2018] [Indexed: 12/27/2022] Open
Abstract
Behavioral comorbidities (depression, anxiety, fatigue, cognitive disturbances, and neuropathic pain) are prevalent in cancer patients and survivors. These mental and neurological health issues reduce quality-of-life, which is a significant societal concern given the increasing rates of long-term survival after various cancers. Hypothesized causes of behavioral comorbidities with cancer include tumor biology, stress associated with the cancer experience, and cancer treatments. A relatively recent leading mechanism by which these causes contribute to changes in neurobiology that underlie behavior is inflammation. Indeed, both basic and clinical research indicates that peripheral inflammation leads to central inflammation and behavioral changes in other illness contexts. Given the limitations of assessing neuroimmunology in clinical populations, this review primarily synthesizes evidence of neuroimmune and neuroinflammatory changes due to two components of cancer (tumor biology and cancer treatments) that are associated with altered affective-like or cognitive behaviors in rodents. Specifically, alterations in microglia, neuroinflammation, and immune trafficking to the brain are compiled in models of tumors, chemotherapy, and/or radiation. Evidence-based neuronal mechanisms by which these neuroimmune changes may lead to changes in behavior are proposed. Finally, converging evidence in clinical cancer populations is discussed.
Collapse
Affiliation(s)
- Jessica C Santos
- Department of Basic and Applied Immunology, School of Medicine of Ribeirao Preto, University of Sao Paulo, Sao Paulo, Brazil
| | - Leah M Pyter
- Departments of Psychiatry and Behavioral Health and Neuroscience, The Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|