Role of the Hypoxic-Secretome in Seed and Soil Metastatic Preparation
Abstract
:Simple Summary
Abstract
1. Introduction
2. Hypoxic Secretome
2.1. Lysyl Oxidase
2.2. VEGF
2.3. TGF-β
2.4. CAIX
2.5. S100A8 and S100A9
2.6. IL-6
2.7. Other Molecules
3. Exosomes
3.1. Effect of Hypoxic Exosomes on Stromal and Immune Cells
3.2. Effect of Hypoxic Exosomes from Stromal Cells on Tumor Cells
3.3. Effect of Hypoxic Exosomes from Stromal Cells on Stromal Cells
4. Effect of Hypoxia on Cellular Components of the Premetastatic Niche
4.1. Effect of Hypoxia on Cellular Senescence and SASP Induction
4.2. Effect of Hypoxia on CSCs
5. Conclusions
Author Contributions
Funding
Conflicts of Interest
Abbreviations
AKT | Protein Kinase B/AKT |
APAF1 | Apoptosis Protease-Activating Factor-1 |
BMDCs | Bone Marrow-Derived Cells |
BMP | Bone Morphogenetic Protein |
CAFs | Cancer Associated Fibroblasts |
CAIX | Carbonic Anhydrase IX |
CAMs | Cell Adhesion Molecules |
CCL9 | Chemokine (C-C motif) Ligand 9 |
CCR1 | C-C Motif Chemokine Receptor 1 |
CKB | Creatine Kinase Brain-type |
CSCs | Cancer Stem Cells |
CTCs | Circulating Tumor Cells |
CXCL | Chemokine (C-X-C motif) Ligand 20 |
CXCR | C-X-C Chemokine Receptor |
ECM | Extracellular Matrix |
EMT | Epithelial to Mesenchymal Transition |
EV | Extracellular Vesicles |
FGF | Fibroblast Growth Factor |
FIH-1 | Factor Inhibiting HIF-1 |
FN | Fibronectin |
GBM | Glioblastoma Multiforme |
G-CSF | Granulocyte Colony Stimulating Factor |
GM-CSF | Granulocyte and Monocyte Colony Stimulating Factor |
GRP78 | 78-kDA glucose-regulated protein |
HIF | Hypoxia-Inducible Factor |
HSP | Heat Shock Protein |
IFN | Interferon |
IL | Interleukine |
LECs | Lymphatic Endothelial Cells |
LOX | Lysyl Oxidase |
MDSCs | Myeloid-Derived Suppressor Cells |
MMP | Matrix Metalloproteinase |
MPVECs | Microvascular Endothelial Cells |
NK | Natural Killer |
PIGF | Placenta Growth Factor |
PMN | Pre Metastatic Niche |
PTHrP | Parathyroid Hormone-related Protein |
ROS | Reactive Oxygen Species |
SAA | Serum Amyloid A Protein |
SASP | Senescence Associated Secretory Phenotype |
Src | Steroid receptor coactivator |
TAMs | Tumor Associated Macrophages |
TDEs | Tumor Derived Exosomes |
TDVs | Tumor Derived Vesicles |
TGF-β | Tumor Growth Factor beta |
TLR | Toll Like Receptor |
TNFα | Tumor Necrosis Factor alpha |
UTR | Untranslated Region |
VEGF | Vascular Endothelial Growth Factor |
VEGFR | Vascular Endothelial Growth Factor Receptor |
VLA4 | Very Late Antigen 4 |
References
- Majidpoor, J.; Mortezaee, K. Steps in metastasis: An updated review. Med. Oncol. 2021, 38, 3. [Google Scholar] [CrossRef] [PubMed]
- Paget, S. The distribution of secondary growths in cancer of the breast. The Lancet 1889, 133, 571–573. [Google Scholar] [CrossRef] [Green Version]
- Kim, M.-Y.; Oskarsson, T.; Acharyya, S.; Nguyen, D.X.; Zhang, X.H.-F.; Norton, L.; Massagué, J. Tumor Self-Seeding by Circulating Cancer Cells. Cell 2009, 139, 1315–1326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Luo, W. Nasopharyngeal Carcinoma Ecology Theory: Cancer as Multidimensional Spatiotemporal “Unity of Ecology and Evolution” Pathological Ecosystem. Preprints 2022, 2022100226. [Google Scholar] [CrossRef]
- Chan, C.Y.-K.; Yuen, V.W.-H.; Wong, C.C.-L. Hypoxia and the Metastatic Niche. In Hypoxia and Cancer Metastasis; Springer: Cham, Switzerland, 2019; pp. 97–112. [Google Scholar]
- Chang, J.; Erler, J. Hypoxia-Mediated Metastasis. Adv. Exp. Med. Biol. 2014, 772, 55–81. [Google Scholar] [CrossRef]
- Araos, J.; Sleeman, J.P.; Garvalov, B.K. The role of hypoxic signalling in metastasis: Towards translating knowledge of basic biology into novel anti-tumour strategies. Clin. Exp. Metastasis 2018, 35, 563–599. [Google Scholar] [CrossRef]
- Chin, A.R.; Wang, S.E. Cancer Tills the Premetastatic Field: Mechanistic Basis and Clinical Implications. Clin. Cancer Res. 2016, 22, 3725–3733. [Google Scholar] [CrossRef] [Green Version]
- Chenau, J.; Michelland, S.; Seve, M. Le sécrétome: Définitions et intérêt biomédical. La Rev. Médecine Interne 2008, 29, 606–608. [Google Scholar] [CrossRef]
- Liu, Y.; Ciotti, G.E.; Eisinger-Mathason, T.S.K. Hypoxia and the Tumor Secretome. In Hypoxia and Cancer Metastasis; Springer: Cham, Switzerland, 2019; pp. 57–69. [Google Scholar]
- Wong, C.C.-L.; Tse, A.P.-W.; Huang, Y.-P.; Zhu, Y.-T.; Chiu, D.K.-C.; Lai, R.K.-H.; Au, S.L.-K.; Kai, A.K.-L.; Lee, J.M.-F.; Wei, L.L.; et al. Lysyl oxidase-like 2 is critical to tumor microenvironment and metastatic niche formation in hepatocellular carcinoma. Hepatology 2014, 60, 1645–1658. [Google Scholar] [CrossRef]
- Erler, J.T.; Bennewith, K.L.; Nicolau, M.; Dornhöfer, N.; Kong, C.; Le, Q.-T.; Chi, J.-T.A.; Jeffrey, S.S.; Giaccia, A.J. Lysyl oxidase is essential for hypoxia-induced metastasis. Nature 2006, 440, 1222–1226. [Google Scholar] [CrossRef]
- Sion, A.M.; Figg, W.D. Lysyl oxidase (LOX) and hypoxia-induced metastases. Cancer Biol. Ther. 2006, 5, 909–911. [Google Scholar] [CrossRef]
- Cox, T.R.; Gartland, A.; Erler, J.T. Lysyl Oxidase, a Targetable Secreted Molecule Involved in Cancer Metastasis. Cancer Res. 2016, 76, 188–192. [Google Scholar] [CrossRef] [Green Version]
- Joshi, H.P.; Subramanian, I.V.; Schnettler, E.K.; Ghosh, G.; Rupaimoole, R.; Evans, C.; Saluja, M.; Jing, Y.; Cristina, I.; Roy, S.; et al. Dynamin 2 along with microRNA-199a reciprocally regulate hypoxia-inducible factors and ovarian cancer metastasis. Proc. Natl. Acad. Sci. USA 2014, 111, 5331–5336. [Google Scholar] [CrossRef] [Green Version]
- Ji, F.; Wang, Y.; Qiu, L.; Li, S.; Zhu, J.; Liang, Z.; Wan, Y.; Di, W. Hypoxia inducible factor 1α-mediated LOX expression correlates with migration and invasion in epithelial ovarian cancer. Int. J. Oncol. 2013, 42, 1578–1588. [Google Scholar] [CrossRef] [Green Version]
- Han, Y.-L.; Chen, L.; Qin, R.; Wang, G.-Q.; Lin, X.-H.; Dai, G.-H. Lysyl oxidase and hypoxia-inducible factor 1α: Biomarkers of gastric cancer. World J. Gastroenterol. 2019, 25, 1828–1839. [Google Scholar] [CrossRef]
- Umezaki, N.; Nakagawa, S.; Yamashita, Y.; Kitano, Y.; Arima, K.; Miyata, T.; Hiyoshi, Y.; Okabe, H.; Nitta, H.; Hayashi, H.; et al. Lysyl oxidase induces epithelial-mesenchymal transition and predicts intrahepatic metastasis of hepatocellular carcinoma. Cancer Sci. 2019, 110, 2033–2043. [Google Scholar] [CrossRef]
- Dai, Z.; Liu, T.; Liu, G.; Deng, Z.; Yu, P.; Wang, B.; Cen, B.; Guo, L.; Zhang, J. Identification of Clinical and Tumor Microenvironment Characteristics of Hypoxia-Related Risk Signature in Lung Adenocarcinoma. Front. Mol. Biosci. 2021, 8, 757421. [Google Scholar] [CrossRef]
- Wong, C.C.-L.; Zhang, H.; Gilkes, D.M.; Chen, J.; Wei, H.; Chaturvedi, P.; Hubbi, M.E.; Semenza, G.L. Inhibitors of hypoxia-inducible factor 1 block breast cancer metastatic niche formation and lung metastasis. J. Mol. Med. 2012, 90, 803–815. [Google Scholar] [CrossRef] [Green Version]
- Urooj, T.; Wasim, B.; Mushtaq, S.; Shah, S.N.N.; Shah, M. Cancer Cell-derived Secretory Factors in Breast Cancer-associated Lung Metastasis: Their Mechanism and Future Prospects. Curr. Cancer Drug Targets 2020, 20, 168–186. [Google Scholar] [CrossRef]
- Wang, Y.; Ma, J.; Shen, H.; Wang, C.; Sun, Y.; Howell, S.B.; Lin, X. Reactive oxygen species promote ovarian cancer progression via the HIF-1α/LOX/E-cadherin pathway. Oncol. Rep. 2014, 32, 2150–2158. [Google Scholar] [CrossRef]
- Joo, Y.N.; Jin, H.; Eun, S.Y.; Park, S.W.; Chang, K.C.; Kim, H.J. P2Y2R activation by nucleotides released from the highly metastatic breast cancer cell contributes to pre-metastatic niche formation by mediating lysyl oxidase secretion, collagen crosslinking, and monocyte recruitment. Oncotarget 2014, 5, 9322–9334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, V.; Davis, D.A.; Yarchoan, R. Identification of functional hypoxia inducible factor response elements in the human lysyl oxidase gene promoter. Biochem. Biophys. Res. Commun. 2017, 490, 480–485. [Google Scholar] [CrossRef] [PubMed]
- Erler, J.T.; Bennewith, K.L.; Cox, T.R.; Lang, G.; Bird, D.; Koong, A.; Le, Q.-T.; Giaccia, A.J. Hypoxia-Induced Lysyl Oxidase Is a Critical Mediator of Bone Marrow Cell Recruitment to form the Premetastatic Niche. Cancer Cell 2009, 15, 35–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Semenza, G.L. Cancer–stromal cell interactions mediated by hypoxia-inducible factors promote angiogenesis, lymphangiogenesis, and metastasis. Oncogene 2013, 32, 4057–4063. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Natarajan, S.; Foreman, K.M.; Soriano, M.I.; Rossen, N.S.; Shehade, H.; Fregoso, D.R.; Eggold, J.T.; Krishnan, V.; Dorigo, O.; Krieg, A.J.; et al. Collagen Remodeling in the Hypoxic Tumor-Mesothelial Niche Promotes Ovarian Cancer Metastasis. Cancer Res. 2019, 79, 2271–2284. [Google Scholar] [CrossRef] [Green Version]
- Wong, C.C.-L.; Gilkes, D.M.; Zhang, H.; Chen, J.; Wei, H.; Chaturvedi, P.; Fraley, S.I.; Wong, C.-M.; Khoo, U.-S.; Ng, I.O.-L.; et al. Hypoxia-inducible factor 1 is a master regulator of breast cancer metastatic niche formation. Proc. Natl. Acad. Sci. USA 2011, 108, 16369–16374. [Google Scholar] [CrossRef] [Green Version]
- Baker, A.-M.; Bird, D.; Lang, G.; Cox, T.R.; Erler, J.T. Lysyl oxidase enzymatic function increases stiffness to drive colorectal cancer progression through FAK. Oncogene 2013, 32, 1863–1868. [Google Scholar] [CrossRef] [Green Version]
- Zhu, G.; Wang, L.; Meng, W.; Lu, S.; Cao, B.; Liang, X.; He, C.; Hao, Y.; Du, X.; Wang, X.; et al. LOXL2-enriched small extracellular vesicles mediate hypoxia-induced premetastatic niche and indicates poor outcome of head and neck cancer. Theranostics 2021, 11, 9198–9216. [Google Scholar] [CrossRef]
- Todd, V.M.; Johnson, R.W. Hypoxia in bone metastasis and osteolysis. Cancer Lett. 2020, 489, 144–154. [Google Scholar] [CrossRef]
- Li, Q.; Zhu, C.-C.; Ni, B.; Zhang, Z.-Z.; Jiang, S.-H.; Hu, L.-P.; Wang, X.; Zhang, X.-X.; Huang, P.-Q.; Yang, Q.; et al. Lysyl oxidase promotes liver metastasis of gastric cancer via facilitating the reciprocal interactions between tumor cells and cancer associated fibroblasts. EBioMedicine 2019, 49, 157–171. [Google Scholar] [CrossRef]
- Yang, Y.-L.; Tsai, M.-C.; Chang, Y.-H.; Wang, C.-C.; Chu, P.-Y.; Lin, H.-Y.; Huang, Y.-H. MIR29A Impedes Metastatic Behaviors in Hepatocellular Carcinoma via Targeting LOX, LOXL2, and VEGFA. Int. J. Mol. Sci. 2021, 22, 6001. [Google Scholar] [CrossRef]
- Miller, B.W.; Morton, J.P.; Pinese, M.; Saturno, G.; Jamieson, N.B.; McGhee, E.; Timpson, P.; Leach, J.; McGarry, L.; Shanks, E.; et al. Targeting the LOX/hypoxia axis reverses many of the features that make pancreatic cancer deadly: Inhibition of LOX abrogates metastasis and enhances drug efficacy. EMBO Mol. Med. 2015, 7, 1063–1076. [Google Scholar] [CrossRef]
- Santhanam, A.N.; Baker, A.R.; Hegamyer, G.; Kirschmann, D.A.; Colburn, N.H. Pdcd4 repression of lysyl oxidase inhibits hypoxia-induced breast cancer cell invasion. Oncogene 2010, 29, 3921–3932. [Google Scholar] [CrossRef] [Green Version]
- Yang, X.; Li, S.; Li, W.; Chen, J.; Xiao, X.; Wang, Y.; Yan, G.; Chen, L. Inactivation of lysyl oxidase by β-aminopropionitrile inhibits hypoxia-induced invasion and migration of cervical cancer cells. Oncol. Rep. 2013, 29, 541–548. [Google Scholar] [CrossRef] [Green Version]
- Salvador, F.; Martin, A.; López-Menéndez, C.; Moreno-Bueno, G.; Santos, V.; Vázquez-Naharro, A.; Santamaria, P.G.; Morales, S.; Dubus, P.R.; Muinelo-Romay, L.; et al. Lysyl Oxidase–like Protein LOXL2 Promotes Lung Metastasis of Breast Cancer. Cancer Res. 2017, 77, 5846–5859. [Google Scholar] [CrossRef] [Green Version]
- Roberts, E.; Cossigny, D.A.F.; Quan, G.M.Y. The Role of Vascular Endothelial Growth Factor in Metastatic Prostate Cancer to the Skeleton. Prostate Cancer 2013, 2013, 418340. [Google Scholar] [CrossRef] [Green Version]
- Wakisaka, N.; Hasegawa, Y.; Yoshimoto, S.; Miura, K.; Shiotani, A.; Yokoyama, J.; Sugasawa, M.; Moriyama-Kita, M.; Endo, K.; Yoshizaki, T. Primary Tumor-Secreted Lymphangiogenic Factors Induce Pre-Metastatic Lymphvascular Niche Formation at Sentinel Lymph Nodes in Oral Squamous Cell Carcinoma. PLoS ONE 2015, 10, e0144056. [Google Scholar] [CrossRef]
- Carano, R.A.; Filvaroff, E.H. Angiogenesis and bone repair. Drug Discov. Today 2003, 8, 980–989. [Google Scholar] [CrossRef]
- Dai, J.; Kitagawa, Y.; Zhang, J.; Yao, Z.; Mizokami, A.; Cheng, S.; Nör, J.; McCauley, L.K.; Taichman, R.S.; Keller, E.T. Vascular Endothelial Growth Factor Contributes to the Prostate Cancer-Induced Osteoblast Differentiation Mediated by Bone Morphogenetic Protein. Cancer Res. 2004, 64, 994–999. [Google Scholar] [CrossRef] [Green Version]
- Kitagawa, Y.; Dai, J.; Zhang, J.; Keller, J.M.; Nor, J.; Yao, Z.; Keller, E.T. Vascular Endothelial Growth Factor Contributes to Prostate Cancer–Mediated Osteoblastic Activity. Cancer Res. 2005, 65, 10921–10929. [Google Scholar] [CrossRef]
- Kaplan, R.N.; Riba, R.D.; Zacharoulis, S.; Bramley, A.H.; Vincent, L.; Costa, C.; MacDonald, D.D.; Jin, D.K.; Shido, K.; Kerns, S.A.; et al. VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature 2005, 438, 820–827. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Van Der Pluijm, G.; Sijmons, B.; Vloedgraven, H.; Deckers, M.; Papapoulos, S.; Löwik, C. Monitoring Metastatic Behavior of Human Tumor Cells in Mice with Species-Specific Polymerase Chain Reaction: Elevated Expression of Angiogenesis and Bone Resorption Stimulators by Breast Cancer in Bone Metastases. J. Bone Miner. Res. 2001, 16, 1077–1091. [Google Scholar] [CrossRef] [PubMed]
- Weilbaecher, K.N.; Guise, T.A.; McCauley, L.K. Cancer to bone: A fatal attraction. Nat. Rev. Cancer 2011, 11, 411–425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hiratsuka, S.; Watanabe, A.; Sakurai, Y.; Akashi-Takamura, S.; Ishibashi, S.; Miyake, K.; Shibuya, M.; Akira, S.; Aburatani, H.; Maru, Y. The S100A8–serum amyloid A3–TLR4 paracrine cascade establishes a pre-metastatic phase. Nat. Cell Biol. 2008, 10, 1349–1355. [Google Scholar] [CrossRef] [PubMed]
- Srikrishna, G. S100A8 and S100A9: New Insights into Their Roles in Malignancy. J. Innate Immun. 2012, 4, 31–40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xu, W.W.; Li, B.; Guan, X.Y.; Chung, S.K.; Wang, Y.; Yip, Y.L.; Law, S.Y.K.; Chan, K.T.; Lee, N.P.Y.; Chan, K.W.; et al. Cancer cell-secreted IGF2 instigates fibroblasts and bone marrow-derived vascular progenitor cells to promote cancer progression. Nat. Commun. 2017, 8, 14399. [Google Scholar] [CrossRef] [Green Version]
- Hiratsuka, S.; Nakamura, K.; Iwai, S.; Murakami, M.; Itoh, T.; Kijima, H.; Shipley, J.M.; Senior, R.M.; Shibuya, M. MMP9 induction by vascular endothelial growth factor receptor-1 is involved in lung-specific metastasis. Cancer Cell 2002, 2, 289–300. [Google Scholar] [CrossRef] [Green Version]
- Chang, W.; Tsai, Y.; Tsai, Y.; Wu, C.; Chang, K.; Lien, C.; Hung, J.; Hsu, Y.; Kuo, P. Differential expression profiles of the transcriptome in bone marrow-derived cells in lung cancer revealed by next generation sequencing and bioinformatics. Oncol. Lett. 2019, 17, 4341–4350. [Google Scholar] [CrossRef]
- Sterling, J.A.; Edwards, J.R.; Martin, T.J.; Mundy, G.R. Advances in the biology of bone metastasis: How the skeleton affects tumor behavior. Bone 2011, 48, 6–15. [Google Scholar] [CrossRef]
- Chen, J.; De, S.; Brainard, J.; Byzova, T.V. Metastatic Properties of Prostate Cancer Cells are Controlled by VEGF. Cell Commun. Adhes. 2004, 11, 1–11. [Google Scholar] [CrossRef]
- Liu, S.; Jiang, M.; Zhao, Q.; Li, S.; Peng, Y.; Zhang, P.; Han, M. Vascular endothelial growth factor plays a critical role in the formation of the pre-metastatic niche via prostaglandin E2. Oncol. Rep. 2014, 32, 2477–2484. [Google Scholar] [CrossRef] [Green Version]
- Hiratsuka, S.; Watanabe, A.; Aburatani, H.; Maru, Y. Tumour-mediated upregulation of chemoattractants and recruitment of myeloid cells predetermines lung metastasis. Nat. Cell Biol. 2006, 8, 1369–1375. [Google Scholar] [CrossRef]
- Efferth, T.; Leber, M.F. Molecular principles of cancer invasion and metastasis (Review). Int. J. Oncol. 2009, 34, 881–895. [Google Scholar] [CrossRef] [Green Version]
- Li, R.; Qi, Y.; Jiang, M.; Zhang, T.; Wang, H.; Wang, L.; Han, M. Primary tumor-secreted VEGF induces vascular hyperpermeability in premetastatic lung via the occludin phosphorylation/ubiquitination pathway. Mol. Carcinog. 2019, 58, 2316–2326. [Google Scholar] [CrossRef]
- Marvin, D.L.; Heijboer, R.; ten Dijke, P.; Ritsma, L. TGF-β signaling in liver metastasis. Clin. Transl. Med. 2020, 10, e160. [Google Scholar] [CrossRef]
- Ayabe, H.; Anada, T.; Kamoya, T.; Sato, T.; Kimura, M.; Yoshizawa, E.; Kikuchi, S.; Ueno, Y.; Sekine, K.; Camp, J.G.; et al. Optimal Hypoxia Regulates Human iPSC-Derived Liver Bud Differentiation through Intercellular TGFB Signaling. Stem. Cell Reports 2018, 11, 306–316. [Google Scholar] [CrossRef] [Green Version]
- Mallikarjuna, P.; Zhou, Y.; Landström, M. The Synergistic Cooperation between TGF-β and Hypoxia in Cancer and Fibrosis. Biomolecules 2022, 12, 635. [Google Scholar] [CrossRef]
- Ashraf, M.A.B.; Zahid, A.; Ashraf, S.; Waquar, S.; Iqbal, S.; Malik, A. Implication of Prophetic Variables and their Impulsive Interplay in CA Prostate Patients Experiencing Osteo-Metastasis. Anticancer. Agents Med. Chem. 2020, 20, 2106–2113. [Google Scholar] [CrossRef]
- Rynne-Vidal, A.; Au-Yeung, C.L.; Jiménez-Heffernan, J.A.; Pérez-Lozano, M.L.; Cremades-Jimeno, L.; Bárcena, C.; Cristóbal-García, I.; Fernández-Chacón, C.; Yeung, T.L.; Mok, S.C.; et al. Mesothelial-to-mesenchymal transition as a possible therapeutic target in peritoneal metastasis of ovarian cancer. J. Pathol. 2017, 242, 140–151. [Google Scholar] [CrossRef] [Green Version]
- Zhu, M.; Zhang, N.; Ma, J.; He, S. Integration of exosomal miR-106a and mesothelial cells facilitates gastric cancer peritoneal dissemination. Cell. Signal. 2022, 91, 110230. [Google Scholar] [CrossRef]
- Mazumdar, A.; Urdinez, J.; Boro, A.; Migliavacca, J.; Arlt, M.J.E.; Muff, R.; Fuchs, B.; Snedeker, J.G.; Gvozdenovic, A. Osteosarcoma-Derived Extracellular Vesicles Induce Lung Fibroblast Reprogramming. Int. J. Mol. Sci. 2020, 21, 5451. [Google Scholar] [CrossRef] [PubMed]
- Li, B.; Xia, Y.; Lv, J.; Wang, W.; Xuan, Z.; Chen, C.; Jiang, T.; Fang, L.; Wang, L.; Li, Z.; et al. miR-151a-3p-rich small extracellular vesicles derived from gastric cancer accelerate liver metastasis via initiating a hepatic stemness-enhancing niche. Oncogene 2021, 40, 6180–6194. [Google Scholar] [CrossRef] [PubMed]
- Yan, H.H.; Jiang, J.; Pang, Y.; Achyut, B.R.; Lizardo, M.; Liang, X.; Hunter, K.; Khanna, C.; Hollander, C.; Yang, L. CCL9 Induced by TGFβ Signaling in Myeloid Cells Enhances Tumor Cell Survival in the Premetastatic Organ. Cancer Res. 2015, 75, 5283–5298. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Batlle, E.; Massagué, J. Transforming Growth Factor-β Signaling in Immunity and Cancer. Immunity 2019, 50, 924–940. [Google Scholar] [CrossRef] [PubMed]
- Tauriello, D.V.F.; Palomo-Ponce, S.; Stork, D.; Berenguer-Llergo, A.; Badia-Ramentol, J.; Iglesias, M.; Sevillano, M.; Ibiza, S.; Cañellas, A.; Hernando-Momblona, X.; et al. TGFβ drives immune evasion in genetically reconstituted colon cancer metastasis. Nature 2018, 554, 538–543. [Google Scholar] [CrossRef] [Green Version]
- Costa-Silva, B.; Aiello, N.M.; Ocean, A.J.; Singh, S.; Zhang, H.; Thakur, B.K.; Becker, A.; Hoshino, A.; Mark, M.T.; Molina, H.; et al. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat. Cell Biol. 2015, 17, 816–826. [Google Scholar] [CrossRef]
- Zhou, W.; Ke, S.Q.; Huang, Z.; Flavahan, W.; Fang, X.; Paul, J.; Wu, L.; Sloan, A.E.; McLendon, R.E.; Li, X.; et al. Periostin secreted by glioblastoma stem cells recruits M2 tumour-associated macrophages and promotes malignant growth. Nat. Cell Biol. 2015, 17, 170–182. [Google Scholar] [CrossRef] [Green Version]
- Kerr, B.A.; Harris, K.S.; Shi, L.; Willey, J.S.; Soto-Pantoja, D.R.; Byzova, T. V Platelet TSP-1 controls prostate cancer-induced osteoclast differentiation and bone marrow-derived cell mobilization through TGFβ-1. Am. J. Clin. Exp. Urol. 2021, 9, 18–31. [Google Scholar]
- Tomar, J.S.; Shen, J. Characterization of Carbonic Anhydrase In Vivo Using Magnetic Resonance Spectroscopy. Int. J. Mol. Sci. 2020, 21, 2442. [Google Scholar] [CrossRef] [Green Version]
- Nolly, M.B.; Vargas, L.A.; Correa, M.V.; Lofeudo, J.M.; Pinilla, A.O.; Rueda, J.O.V.; Guerrero-Gimenez, M.E.; Swenson, E.R.; Damiani, M.T.; Alvarez, B.V. Carbonic anhydrase IX and hypoxia-inducible factor 1 attenuate cardiac dysfunction after myocardial infarction. Pflügers Arch. Eur. J. Physiol. 2021, 473, 1273–1285. [Google Scholar] [CrossRef]
- McDonald, P.C.; Dedhar, S. Carbonic Anhydrase IX (CAIX) as a Mediator of Hypoxia-Induced Stress Response in Cancer Cells; Springer: Dordrecht, The Netherlands, 2014; pp. 255–269. [Google Scholar]
- Ong, C.H.C.; Lee, D.Y.; Lee, B.; Li, H.; Lim, J.C.T.; Lim, J.X.; Yeong, J.P.S.; Lau, H.Y.; Thike, A.A.; Tan, P.H.; et al. Hypoxia-regulated carbonic anhydrase IX (CAIX) protein is an independent prognostic indicator in triple negative breast cancer. Breast Cancer Res. 2022, 24, 38. [Google Scholar] [CrossRef]
- Hedlund, E.-M.E.; McDonald, P.C.; Nemirovsky, O.; Awrey, S.; Jensen, L.D.E.; Dedhar, S. Harnessing Induced Essentiality: Targeting Carbonic Anhydrase IX and Angiogenesis Reduces Lung Metastasis of Triple Negative Breast Cancer Xenografts. Cancers 2019, 11, 1002. [Google Scholar] [CrossRef] [Green Version]
- Schmidt, J.; Oppermann, E.; Blaheta, R.A.; Schreckenbach, T.; Lunger, I.; Rieger, M.A.; Bechstein, W.O.; Holzer, K.; Malkomes, P. Carbonic-anhydrase IX expression is increased in thyroid cancer tissue and represents a potential therapeutic target to eradicate thyroid tumor-initiating cells. Mol. Cell. Endocrinol. 2021, 535, 111382. [Google Scholar] [CrossRef]
- Kalinin, S.; Malkova, A.; Sharonova, T.; Sharoyko, V.; Bunev, A.; Supuran, C.T.; Krasavin, M. Carbonic Anhydrase IX Inhibitors as Candidates for Combination Therapy of Solid Tumors. Int. J. Mol. Sci. 2021, 22, 13405. [Google Scholar] [CrossRef]
- Hsin, M.-C.; Hsieh, Y.-H.; Hsiao, Y.-H.; Chen, P.-N.; Wang, P.-H.; Yang, S.-F. Carbonic Anhydrase IX Promotes Human Cervical Cancer Cell Motility by Regulating PFKFB4 Expression. Cancers 2021, 13, 1174. [Google Scholar] [CrossRef]
- Zhang, X.; Liu, X.; Cui, W.; Zhang, R.; Liu, Y.; Li, Y.; Hao, J. Sohlh2 alleviates malignancy of EOC cells under hypoxia via inhibiting the HIF1α/CA9 signaling pathway. Biol. Chem. 2020, 401, 263–271. [Google Scholar] [CrossRef]
- Pastorekova, S.; Gillies, R.J. The role of carbonic anhydrase IX in cancer development: Links to hypoxia, acidosis, and beyond. Cancer Metastasis Rev. 2019, 38, 65–77. [Google Scholar] [CrossRef]
- Swayampakula, M.; McDonald, P.C.; Vallejo, M.; Coyaud, E.; Chafe, S.C.; Westerback, A.; Venkateswaran, G.; Shankar, J.; Gao, G.; Laurent, E.M.N.; et al. The interactome of metabolic enzyme carbonic anhydrase IX reveals novel roles in tumor cell migration and invadopodia/MMP14-mediated invasion. Oncogene 2017, 36, 6244–6261. [Google Scholar] [CrossRef] [Green Version]
- Yang, J.-S.; Lin, C.-W.; Hsieh, Y.-H.; Chien, M.-H.; Chuang, C.-Y.; Yang, S.-F. Overexpression of carbonic anhydrase IX induces cell motility by activating matrix metalloproteinase-9 in human oral squamous cell carcinoma cells. Oncotarget 2017, 8, 83088–83099. [Google Scholar] [CrossRef] [Green Version]
- Chafe, S.C.; Lou, Y.; Sceneay, J.; Vallejo, M.; Hamilton, M.J.; McDonald, P.C.; Bennewith, K.L.; Möller, A.; Dedhar, S. Carbonic Anhydrase IX Promotes Myeloid-Derived Suppressor Cell Mobilization and Establishment of a Metastatic Niche by Stimulating G-CSF Production. Cancer Res. 2015, 75, 996–1008. [Google Scholar] [CrossRef] [Green Version]
- Kowanetz, M.; Wu, X.; Lee, J.; Tan, M.; Hagenbeek, T.; Qu, X.; Yu, L.; Ross, J.; Korsisaari, N.; Cao, T.; et al. Granulocyte-colony stimulating factor promotes lung metastasis through mobilization of Ly6G+Ly6C+ granulocytes. Proc. Natl. Acad. Sci. USA 2010, 107, 21248–21255. [Google Scholar] [CrossRef] [PubMed]
- Sceneay, J.; Chow, M.T.; Chen, A.; Halse, H.M.; Wong, C.S.F.; Andrews, D.M.; Sloan, E.K.; Parker, B.S.; Bowtell, D.D.; Smyth, M.J.; et al. Primary Tumor Hypoxia Recruits CD11b+/Ly6Cmed/Ly6G+ Immune Suppressor Cells and Compromises NK Cell Cytotoxicity in the Premetastatic Niche. Cancer Res. 2012, 72, 3906–3911. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Horie, K.; Kawakami, K.; Fujita, Y.; Sugaya, M.; Kameyama, K.; Mizutani, K.; Deguchi, T.; Ito, M. Exosomes expressing carbonic anhydrase 9 promote angiogenesis. Biochem. Biophys. Res. Commun. 2017, 492, 356–361. [Google Scholar] [CrossRef] [PubMed]
- Ledaki, I.; McIntyre, A.; Wigfield, S.; Buffa, F.; McGowan, S.; Baban, D.; Li, J.; Harris, A.L. Carbonic anhydrase IX induction defines a heterogeneous cancer cell response to hypoxia and mediates stem cell-like properties and sensitivity to HDAC inhibition. Oncotarget 2015, 6, 19413–19427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Marie-Egyptienne, D.T.; Chaudary, N.; Kalliomäki, T.; Hedley, D.W.; Hill, R.P. Cancer initiating-cells are enriched in the CA9 positive fraction of primary cervix cancer xenografts. Oncotarget 2017, 8, 1392–1404. [Google Scholar] [CrossRef] [Green Version]
- Tomita, T.; Sakurai, Y.; Ishibashi, S.; Maru, Y. Imbalance of Clara cell-mediated homeostatic inflammation is involved in lung metastasis. Oncogene 2011, 30, 3429–3439. [Google Scholar] [CrossRef] [Green Version]
- Lukanidin, E.; Sleeman, J.P. Building the niche: The role of the S100 proteins in metastatic growth. Semin. Cancer Biol. 2012, 22, 216–225. [Google Scholar] [CrossRef]
- Grivennikov, S.I.; Karin, M. Inflammatory cytokines in cancer: Tumour necrosis factor and interleukin 6 take the stage. Ann. Rheum. Dis. 2011, 70, i104–i108. [Google Scholar] [CrossRef]
- Gao, D.; Joshi, N.; Choi, H.; Ryu, S.; Hahn, M.; Catena, R.; Sadik, H.; Argani, P.; Wagner, P.; Vahdat, L.T.; et al. Myeloid Progenitor Cells in the Premetastatic Lung Promote Metastases by Inducing Mesenchymal to Epithelial Transition. Cancer Res. 2012, 72, 1384–1394. [Google Scholar] [CrossRef] [Green Version]
- Yan, H.H.; Pickup, M.; Pang, Y.; Gorska, A.E.; Li, Z.; Chytil, A.; Geng, Y.; Gray, J.W.; Moses, H.L.; Yang, L. Gr-1+CD11b+ Myeloid Cells Tip the Balance of Immune Protection to Tumor Promotion in the Premetastatic Lung. Cancer Res. 2010, 70, 6139–6149. [Google Scholar] [CrossRef] [Green Version]
- Jing, B.; Wang, T.; Sun, B.; Xu, J.; Xu, D.; Liao, Y.; Song, H.; Guo, W.; Li, K.; Hu, M.; et al. IL6/STAT3 Signaling Orchestrates Premetastatic Niche Formation and Immunosuppressive Traits in Lung. Cancer Res. 2020, 80, 784–797. [Google Scholar] [CrossRef]
- Lee, E.; Fertig, E.J.; Jin, K.; Sukumar, S.; Pandey, N.B.; Popel, A.S. Breast cancer cells condition lymphatic endothelial cells within pre-metastatic niches to promote metastasis. Nat. Commun. 2014, 5, 4715. [Google Scholar] [CrossRef] [Green Version]
- Bellido, T.; Borba, V.Z.C.; Roberson, P.; Manolagas, S.C. Activation of the Janus Kinase/STAT (Signal Transducer and Activator of Transcription) Signal Transduction Pathway by Interleukin-6-Type Cytokines Promotes Osteoblast Differentiation*. Endocrinology 1997, 138, 3666–3676. [Google Scholar] [CrossRef]
- Sottnik, J.L.; Keller, E.T. Understanding and Targeting Osteoclastic Activity in Prostate Cancer Bone Metastases. Curr. Mol. Med. 2013, 13, 626–639. [Google Scholar] [CrossRef] [Green Version]
- Yin, J.J.; Pollock, C.B.; Kelly, K. Mechanisms of cancer metastasis to the bone. Cell Res. 2005, 15, 57–62. [Google Scholar] [CrossRef] [Green Version]
- Ibrahim, T.; Flamini, E.; Mercatali, L.; Sacanna, E.; Serra, P.; Amadori, D. Pathogenesis of osteoblastic bone metastases from prostate cancer. Cancer 2010, 116, 1406–1418. [Google Scholar] [CrossRef]
- Rucci, N.; Angelucci, A. Prostate Cancer and Bone: The Elective Affinities. Biomed Res. Int. 2014, 2014, 167035. [Google Scholar] [CrossRef] [Green Version]
- Mizutani, K.; Sud, S.; Pienta, K.J. Prostate cancer promotes CD11b positive cells to differentiate into osteoclasts. J. Cell. Biochem. 2009, 106, 563–569. [Google Scholar] [CrossRef]
- Kim, S.W.; Kim, J.S.; Papadopoulos, J.; Choi, H.J.; He, J.; Maya, M.; Langley, R.R.; Fan, D.; Fidler, I.J.; Kim, S.-J. Consistent interactions between tumor cell IL-6 and macrophage TNF-α enhance the growth of human prostate cancer cells in the bone of nude mouse. Int. Immunopharmacol. 2011, 11, 862–872. [Google Scholar] [CrossRef] [Green Version]
- Jiang, J.; Wang, G.-Z.; Wang, Y.; Huang, H.-Z.; Li, W.-T.; Qu, X.-D. Hypoxia-induced HMGB1 expression of HCC promotes tumor invasiveness and metastasis via regulating macrophage-derived IL-6. Exp. Cell Res. 2018, 367, 81–88. [Google Scholar] [CrossRef]
- Wang, X.; Lee, S.O.; Xia, S.; Jiang, Q.; Luo, J.; Li, L.; Yeh, S.; Chang, C. Endothelial Cells Enhance Prostate Cancer Metastasis via IL-6→Androgen Receptor→TGF-β→MMP-9 Signals. Mol. Cancer Ther. 2013, 12, 1026–1037. [Google Scholar] [CrossRef] [PubMed]
- Manisterski, M.; Golan, M.; Amir, S.; Weisman, Y.; Mabjeesh, N.J. Hypoxia induces PTHrP gene transcription in human cancer cells through the HIF-2α. Cell Cycle 2010, 9, 3723–3729. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rankin, E.B.; Giaccia, A.J. Hypoxic control of metastasis. Science 2016, 352, 175–180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Guo, F.; Wang, Y.; Liu, J.; Mok, S.C.; Xue, F.; Zhang, W. CXCL12/CXCR4: A symbiotic bridge linking cancer cells and their stromal neighbors in oncogenic communication networks. Oncogene 2016, 35, 816–826. [Google Scholar] [CrossRef] [PubMed]
- Jin, F.; Brockmeier, U.; Otterbach, F.; Metzen, E. New Insight into the SDF-1/CXCR4 Axis in a Breast Carcinoma Model: Hypoxia-Induced Endothelial SDF-1 and Tumor Cell CXCR4 Are Required for Tumor Cell Intravasation. Mol. Cancer Res. 2012, 10, 1021–1031. [Google Scholar] [CrossRef] [Green Version]
- Dai, J.; Escara-Wilke, J.; Keller, J.M.; Jung, Y.; Taichman, R.S.; Pienta, K.J.; Keller, E.T. Primary prostate cancer educates bone stroma through exosomal pyruvate kinase M2 to promote bone metastasis. J. Exp. Med. 2019, 216, 2883–2899. [Google Scholar] [CrossRef]
- Greer, S.N.; Metcalf, J.L.; Wang, Y.; Ohh, M. The updated biology of hypoxia-inducible factor. EMBO J. 2012, 31, 2448–2460. [Google Scholar] [CrossRef] [Green Version]
- Oskarsson, T.; Acharyya, S.; Zhang, X.H.-F.; Vanharanta, S.; Tavazoie, S.F.; Morris, P.G.; Downey, R.J.; Manova-Todorova, K.; Brogi, E.; Massagué, J. Breast cancer cells produce tenascin C as a metastatic niche component to colonize the lungs. Nat. Med. 2011, 17, 867–874. [Google Scholar] [CrossRef] [Green Version]
- Loo, J.M.; Scherl, A.; Nguyen, A.; Man, F.Y.; Weinberg, E.; Zeng, Z.; Saltz, L.; Paty, P.B.; Tavazoie, S.F. Extracellular Metabolic Energetics Can Promote Cancer Progression. Cell 2015, 160, 393–406. [Google Scholar] [CrossRef] [Green Version]
- Lee, J.; Yoon, Y.; Lee, S. Hypoxic Preconditioning Promotes the Bioactivities of Mesenchymal Stem Cells via the HIF-1α-GRP78-Akt Axis. Int. J. Mol. Sci. 2017, 18, 1320. [Google Scholar] [CrossRef]
- Ramirez, M.U.; Hernandez, S.R.; Soto-Pantoja, D.R.; Cook, K.L. Endoplasmic Reticulum Stress Pathway, the Unfolded Protein Response, Modulates Immune Function in the Tumor Microenvironment to Impact Tumor Progression and Therapeutic Response. Int. J. Mol. Sci. 2019, 21, 169. [Google Scholar] [CrossRef]
- Chen, L.; Zheng, H.; Yu, X.; Liu, L.; Li, H.; Zhu, H.; Zhang, Z.; Lei, P.; Shen, G. Tumor-Secreted GRP78 Promotes the Establishment of a Pre-metastatic Niche in the Liver Microenvironment. Front. Immunol. 2020, 11, 584458. [Google Scholar] [CrossRef]
- Qiang, J.; He, J.; Tao, Y.-F.; Bao, J.-W.; Zhu, J.-H.; Xu, P. Hypoxia-induced miR-92a regulates p53 signaling pathway and apoptosis by targeting calcium-sensing receptor in genetically improved farmed tilapia (Oreochromis niloticus). PLoS ONE 2020, 15, e0238897. [Google Scholar] [CrossRef]
- Hsu, Y.-L.; Huang, M.-S.; Hung, J.-Y.; Chang, W.-A.; Tsai, Y.-M.; Pan, Y.-C.; Lin, Y.-S.; Tsai, H.-P.; Kuo, P.-L. Bone-marrow-derived cell-released extracellular vesicle miR-92a regulates hepatic pre-metastatic niche in lung cancer. Oncogene 2020, 39, 739–753. [Google Scholar] [CrossRef]
- Kahlert, C.; Kalluri, R. Exosomes in tumor microenvironment influence cancer progression and metastasis. J. Mol. Med. 2013, 91, 431–437. [Google Scholar] [CrossRef] [Green Version]
- Raposo, G.; Nijman, H.W.; Stoorvogel, W.; Liejendekker, R.; Harding, C.V.; Melief, C.J.; Geuze, H.J. B lymphocytes secrete antigen-presenting vesicles. J. Exp. Med. 1996, 183, 1161–1172. [Google Scholar] [CrossRef]
- Aga, M.; Bentz, G.L.; Raffa, S.; Torrisi, M.R.; Kondo, S.; Wakisaka, N.; Yoshizaki, T.; Pagano, J.S.; Shackelford, J. Exosomal HIF1α supports invasive potential of nasopharyngeal carcinoma-associated LMP1-positive exosomes. Oncogene 2014, 33, 4613–4622. [Google Scholar] [CrossRef] [Green Version]
- Shan, Y.; You, B.; Shi, S.; Shi, W.; Zhang, Z.; Zhang, Q.; Gu, M.; Chen, J.; Bao, L.; Liu, D.; et al. Hypoxia-Induced Matrix Metalloproteinase-13 Expression in Exosomes from Nasopharyngeal Carcinoma Enhances Metastases. Cell Death Dis. 2018, 9, 382. [Google Scholar] [CrossRef] [Green Version]
- Li, L.; Li, C.; Wang, S.; Wang, Z.; Jiang, J.; Wang, W.; Li, X.; Chen, J.; Liu, K.; Li, C.; et al. Exosomes Derived from Hypoxic Oral Squamous Cell Carcinoma Cells Deliver miR-21 to Normoxic Cells to Elicit a Prometastatic Phenotype. Cancer Res. 2016, 76, 1770–1780. [Google Scholar] [CrossRef] [Green Version]
- Xue, M.; Chen, W.; Xiang, A.; Wang, R.; Chen, H.; Pan, J.; Pang, H.; An, H.; Wang, X.; Hou, H.; et al. Hypoxic exosomes facilitate bladder tumor growth and development through transferring long non-coding RNA-UCA1. Mol. Cancer 2017, 16, 143. [Google Scholar] [CrossRef] [Green Version]
- Wang, Y.; Yi, J.; Chen, X.; Zhang, Y.; Xu, M.; Yang, Z. The regulation of cancer cell migration by lung cancer cell-derived exosomes through TGF-β and IL-10. Oncol. Lett. 2016, 11, 1527–1530. [Google Scholar] [CrossRef] [PubMed]
- Ramteke, A.; Ting, H.; Agarwal, C.; Mateen, S.; Somasagara, R.; Hussain, A.; Graner, M.; Frederick, B.; Agarwal, R.; Deep, G. Exosomes secreted under hypoxia enhance invasiveness and stemness of prostate cancer cells by targeting adherens junction molecules. Mol. Carcinog. 2015, 54, 554–565. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, T.; Gilkes, D.M.; Takano, N.; Xiang, L.; Luo, W.; Bishop, C.J.; Chaturvedi, P.; Green, J.J.; Semenza, G.L. Hypoxia-inducible factors and RAB22A mediate formation of microvesicles that stimulate breast cancer invasion and metastasis. Proc. Natl. Acad. Sci. USA 2014, 111, E3234–E3242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ngora, H.; Galli, U.M.; Miyazaki, K.; Zöller, M. Membrane-Bound and Exosomal Metastasis-Associated C4.4A Promotes Migration by Associating with the α6β4 Integrin and MT1-MMP. Neoplasia 2012, 14, 95-IN2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gutwein, P.; Stoeck, A.; Riedle, S.; Gast, D.; Runz, S.; Condon, T.P.; Marmé, A.; Phong, M.-C.; Linderkamp, O.; Skorokhod, A.; et al. Cleavage of L1 in Exosomes and Apoptotic Membrane Vesicles Released from Ovarian Carcinoma Cells. Clin. Cancer Res. 2005, 11, 2492–2501. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Peinado, H.; Lavotshkin, S.; Lyden, D. The secreted factors responsible for pre-metastatic niche formation: Old sayings and new thoughts. Semin. Cancer Biol. 2011, 21, 139–146. [Google Scholar] [CrossRef]
- Peinado, H.; Alečković, M.; Lavotshkin, S.; Matei, I.; Costa-Silva, B.; Moreno-Bueno, G.; Hergueta-Redondo, M.; Williams, C.; García-Santos, G.; Ghajar, C.M.; et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat. Med. 2012, 18, 883–891. [Google Scholar] [CrossRef] [Green Version]
- Qian, D.; Xie, Y.; Huang, M.; Gu, J. Tumor-derived exosomes in hypoxic microenvironment: Release mechanism, biological function and clinical application. J. Cancer 2022, 13, 1685–1694. [Google Scholar] [CrossRef]
- Chen, X.; Zhou, J.; Li, X.; Wang, X.; Lin, Y.; Wang, X. Exosomes derived from hypoxic epithelial ovarian cancer cells deliver microRNAs to macrophages and elicit a tumor-promoted phenotype. Cancer Lett. 2018, 435, 80–91. [Google Scholar] [CrossRef]
- Umezu, T.; Tadokoro, H.; Azuma, K.; Yoshizawa, S.; Ohyashiki, K.; Ohyashiki, J.H. Exosomal miR-135b shed from hypoxic multiple myeloma cells enhances angiogenesis by targeting factor-inhibiting HIF-1. Blood 2014, 124, 3748–3757. [Google Scholar] [CrossRef] [Green Version]
- Mace, T.A.; Collins, A.L.; Wojcik, S.E.; Croce, C.M.; Lesinski, G.B.; Bloomston, M. Hypoxia induces the overexpression of microRNA-21 in pancreatic cancer cells. J. Surg. Res. 2013, 184, 855–860. [Google Scholar] [CrossRef]
- King, H.W.; Michael, M.Z.; Gleadle, J.M. Hypoxic enhancement of exosome release by breast cancer cells. BMC Cancer 2012, 12, 421. [Google Scholar] [CrossRef] [Green Version]
- Dorayappan, K.D.P.; Wanner, R.; Wallbillich, J.J.; Saini, U.; Zingarelli, R.; Suarez, A.A.; Cohn, D.E.; Selvendiran, K. Hypoxia-induced exosomes contribute to a more aggressive and chemoresistant ovarian cancer phenotype: A novel mechanism linking STAT3/Rab proteins. Oncogene 2018, 37, 3806–3821. [Google Scholar] [CrossRef]
- Panigrahi, G.K.; Praharaj, P.P.; Peak, T.C.; Long, J.; Singh, R.; Rhim, J.S.; Abd Elmageed, Z.Y.; Deep, G. Hypoxia-induced exosome secretion promotes survival of African-American and Caucasian prostate cancer cells. Sci. Rep. 2018, 8, 3853. [Google Scholar] [CrossRef]
- Wang, Z.; Jin, N.; Ganguli, S.; Swartz, D.R.; Li, L.; Rhoades, R.A. Rho-Kinase Activation Is Involved in Hypoxia-Induced Pulmonary Vasoconstriction. Am. J. Respir. Cell Mol. Biol. 2001, 25, 628–635. [Google Scholar] [CrossRef]
- Pasquet, J.-M.; Toti, F.; Nurden, A.T.; Dachary-Prigent, J. Procoagulant activity and active calpain in platelet-derived microparticles. Thromb. Res. 1996, 82, 509–522. [Google Scholar] [CrossRef]
- Novgorodov, S.A.; Gudz, T.I. Ceramide and Mitochondria in Ischemia/Reperfusion. J. Cardiovasc. Pharmacol. 2009, 53, 198–208. [Google Scholar] [CrossRef] [Green Version]
- Meng, W.; Hao, Y.; He, C.; Li, L.; Zhu, G. Exosome-orchestrated hypoxic tumor microenvironment. Mol. Cancer 2019, 18, 57. [Google Scholar] [CrossRef] [Green Version]
- Ludwig, N.; Whiteside, T.L. Potential roles of tumor-derived exosomes in angiogenesis. Expert Opin. Ther. Targets 2018, 22, 409–417. [Google Scholar] [CrossRef]
- Park, J.E.; Tan, H.S.; Datta, A.; Lai, R.C.; Zhang, H.; Meng, W.; Lim, S.K.; Sze, S.K. Hypoxic Tumor Cell Modulates Its Microenvironment to Enhance Angiogenic and Metastatic Potential by Secretion of Proteins and Exosomes. Mol. Cell. Proteomics 2010, 9, 1085–1099. [Google Scholar] [CrossRef] [Green Version]
- Tadokoro, H.; Umezu, T.; Ohyashiki, K.; Hirano, T.; Ohyashiki, J.H. Exosomes Derived from Hypoxic Leukemia Cells Enhance Tube Formation in Endothelial Cells. J. Biol. Chem. 2013, 288, 34343–34351. [Google Scholar] [CrossRef] [PubMed]
- Hsu, Y.-L.; Hung, J.-Y.; Chang, W.-A.; Lin, Y.-S.; Pan, Y.-C.; Tsai, P.-H.; Wu, C.-Y.; Kuo, P.-L. Hypoxic lung cancer-secreted exosomal miR-23a increased angiogenesis and vascular permeability by targeting prolyl hydroxylase and tight junction protein ZO-1. Oncogene 2017, 36, 4929–4942. [Google Scholar] [CrossRef] [PubMed]
- Paggetti, J.; Haderk, F.; Seiffert, M.; Janji, B.; Distler, U.; Ammerlaan, W.; Kim, Y.J.; Adam, J.; Lichter, P.; Solary, E.; et al. Exosomes released by chronic lymphocytic leukemia cells induce the transition of stromal cells into cancer-associated fibroblasts. Blood 2015, 126, 1106–1117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, J.; Wu, S.; Zheng, X.; Zheng, P.; Fu, Y.; Wu, C.; Lu, B.; Ju, J.; Jiang, J. Immune suppressed tumor microenvironment by exosomes derived from gastric cancer cells via modulating immune functions. Sci. Rep. 2020, 10, 14749. [Google Scholar] [CrossRef] [PubMed]
- Yang, L.; Zhang, W.; Wang, Y.; Zou, T.; Zhang, B.; Xu, Y.; Pang, T.; Hu, Q.; Chen, M.; Wang, L.; et al. Hypoxia-induced miR-214 expression promotes tumour cell proliferation and migration by enhancing the Warburg effect in gastric carcinoma cells. Cancer Lett. 2018, 414, 44–56. [Google Scholar] [CrossRef]
- Ning, T.; Li, J.; He, Y.; Zhang, H.; Wang, X.; Deng, T.; Liu, R.; Li, H.; Bai, M.; Fan, Q.; et al. Exosomal miR-208b related with oxaliplatin resistance promotes Treg expansion in colorectal cancer. Mol. Ther. 2021, 29, 2723–2736. [Google Scholar] [CrossRef]
- Ye, S.-B.; Zhang, H.; Cai, T.-T.; Liu, Y.-N.; Ni, J.-J.; He, J.; Peng, J.-Y.; Chen, Q.-Y.; Mo, H.-Y.; Jun-Cui; et al. Exosomal miR-24-3p impedes T-cell function by targeting FGF11 and serves as a potential prognostic biomarker for nasopharyngeal carcinoma. J. Pathol. 2016, 240, 329–340. [Google Scholar] [CrossRef]
- Li, L.; Cao, B.; Liang, X.; Lu, S.; Luo, H.; Wang, Z.; Wang, S.; Jiang, J.; Lang, J.; Zhu, G. Microenvironmental oxygen pressure orchestrates an anti- and pro-tumoral γδ T cell equilibrium via tumor-derived exosomes. Oncogene 2019, 38, 2830–2843. [Google Scholar] [CrossRef]
- Rong, L.; Li, R.; Li, S.; Luo, R. Immunosuppression of breast cancer cells mediated by transforming growth factor-β in exosomes from cancer cells. Oncol. Lett. 2016, 11, 500–504. [Google Scholar] [CrossRef] [Green Version]
- Wang, X.; Luo, G.; Zhang, K.; Cao, J.; Huang, C.; Jiang, T.; Liu, B.; Su, L.; Qiu, Z. Hypoxic Tumor-Derived Exosomal miR-301a Mediates M2 Macrophage Polarization via PTEN/PI3Kγ to Promote Pancreatic Cancer Metastasis. Cancer Res. 2018, 78, 4586–4598. [Google Scholar] [CrossRef] [Green Version]
- Hsu, Y.-L.; Hung, J.-Y.; Chang, W.-A.; Jian, S.-F.; Lin, Y.-S.; Pan, Y.-C.; Wu, C.-Y.; Kuo, P.-L. Hypoxic Lung-Cancer-Derived Extracellular Vesicle MicroRNA-103a Increases the Oncogenic Effects of Macrophages by Targeting PTEN. Mol. Ther. 2018, 26, 568–581. [Google Scholar] [CrossRef]
- Baginska, J.; Viry, E.; Paggetti, J.; Medves, S.; Berchem, G.; Moussay, E.; Janji, B. The Critical Role of the Tumor Microenvironment in Shaping Natural Killer Cell-Mediated Anti-Tumor Immunity. Front. Immunol. 2013, 4, 490. [Google Scholar] [CrossRef] [Green Version]
- Wen, S.W.; Sceneay, J.; Lima, L.G.; Wong, C.S.F.; Becker, M.; Krumeich, S.; Lobb, R.J.; Castillo, V.; Wong, K.N.; Ellis, S.; et al. The Biodistribution and Immune Suppressive Effects of Breast Cancer–Derived Exosomes. Cancer Res. 2016, 76, 6816–6827. [Google Scholar] [CrossRef] [Green Version]
- Berchem, G.; Noman, M.Z.; Bosseler, M.; Paggetti, J.; Baconnais, S.; Le cam, E.; Nanbakhsh, A.; Moussay, E.; Mami-Chouaib, F.; Janji, B.; et al. Hypoxic tumor-derived microvesicles negatively regulate NK cell function by a mechanism involving TGF-β and miR23a transfer. Oncoimmunology 2016, 5, e1062968. [Google Scholar] [CrossRef] [Green Version]
- Zhang, P.-F.; Gao, C.; Huang, X.-Y.; Lu, J.-C.; Guo, X.-J.; Shi, G.-M.; Cai, J.-B.; Ke, A.-W. Cancer cell-derived exosomal circUHRF1 induces natural killer cell exhaustion and may cause resistance to anti-PD1 therapy in hepatocellular carcinoma. Mol. Cancer 2020, 19, 110. [Google Scholar] [CrossRef]
- Chalmin, F.; Ladoire, S.; Mignot, G.; Vincent, J.; Bruchard, M.; Remy-Martin, J.-P.; Boireau, W.; Rouleau, A.; Simon, B.; Lanneau, D.; et al. Membrane-associated Hsp72 from tumor-derived exosomes mediates STAT3-dependent immunosuppressive function of mouse and human myeloid-derived suppressor cells. J. Clin. Investig. 2010, 120, 457–471. [Google Scholar] [CrossRef]
- Xiang, X.; Liu, Y.; Zhuang, X.; Zhang, S.; Michalek, S.; Taylor, D.D.; Grizzle, W.; Zhang, H.-G. TLR2-Mediated Expansion of MDSCs Is Dependent on the Source of Tumor Exosomes. Am. J. Pathol. 2010, 177, 1606–1610. [Google Scholar] [CrossRef]
- Salem, K.Z.; Moschetta, M.; Sacco, A.; Imberti, L.; Rossi, G.; Ghobrial, I.M.; Manier, S.; Roccaro, A.M. Exosomes in Tumor Angiogenesis. Methods Mol. Biol. 2016, 1464, 25–34. [Google Scholar] [CrossRef]
- Yang, M.; Chen, J.; Su, F.; Yu, B.; Su, F.; Lin, L.; Liu, Y.; Huang, J.-D.; Song, E. Microvesicles secreted by macrophages shuttle invasion-potentiating microRNAs into breast cancer cells. Mol. Cancer 2011, 10, 117. [Google Scholar] [CrossRef] [Green Version]
- Zheng, P.; Luo, Q.; Wang, W.; Li, J.; Wang, T.; Wang, P.; Chen, L.; Zhang, P.; Chen, H.; Liu, Y.; et al. Tumor-associated macrophages-derived exosomes promote the migration of gastric cancer cells by transfer of functional Apolipoprotein E. Cell Death Dis. 2018, 9, 434. [Google Scholar] [CrossRef] [Green Version]
- Au Yeung, C.L.; Co, N.-N.; Tsuruga, T.; Yeung, T.-L.; Kwan, S.-Y.; Leung, C.S.; Li, Y.; Lu, E.S.; Kwan, K.; Wong, K.-K.; et al. Exosomal transfer of stroma-derived miR21 confers paclitaxel resistance in ovarian cancer cells through targeting APAF1. Nat. Commun. 2016, 7, 11150. [Google Scholar] [CrossRef] [PubMed]
- Ismail, N.; Wang, Y.; Dakhlallah, D.; Moldovan, L.; Agarwal, K.; Batte, K.; Shah, P.; Wisler, J.; Eubank, T.D.; Tridandapani, S.; et al. Macrophage microvesicles induce macrophage differentiation and miR-223 transfer. Blood 2013, 121, 984–995. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rodier, F.; Coppé, J.-P.; Patil, C.K.; Hoeijmakers, W.A.M.; Muñoz, D.P.; Raza, S.R.; Freund, A.; Campeau, E.; Davalos, A.R.; Campisi, J. Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion. Nat. Cell Biol. 2009, 11, 973–979. [Google Scholar] [CrossRef] [PubMed]
- Faget, D.V.; Ren, Q.; Stewart, S.A. Unmasking senescence: Context-dependent effects of SASP in cancer. Nat. Rev. Cancer 2019, 19, 439–453. [Google Scholar] [CrossRef]
- Mortezaee, K.; Majidpoor, J. Key promoters of tumor hallmarks. Int. J. Clin. Oncol. 2022, 27, 45–58. [Google Scholar] [CrossRef]
- Davalos, A.R.; Coppe, J.-P.; Campisi, J.; Desprez, P.-Y. Senescent cells as a source of inflammatory factors for tumor progression. Cancer Metastasis Rev. 2010, 29, 273–283. [Google Scholar] [CrossRef] [Green Version]
- Debacq-Chainiaux, F.; Erusalimsky, J.D.; Campisi, J.; Toussaint, O. Protocols to detect senescence-associated beta-galactosidase (SA-βgal) activity, a biomarker of senescent cells in culture and in vivo. Nat. Protoc. 2009, 4, 1798–1806. [Google Scholar] [CrossRef]
- Coppé, J.-P.; Patil, C.K.; Rodier, F.; Sun, Y.; Muñoz, D.P.; Goldstein, J.; Nelson, P.S.; Desprez, P.-Y.; Campisi, J. Senescence-Associated Secretory Phenotypes Reveal Cell-Nonautonomous Functions of Oncogenic RAS and the p53 Tumor Suppressor. PLoS Biol. 2008, 6, e301. [Google Scholar] [CrossRef]
- Garfinkel, S.; Brown, S.; Wessendorf, J.H.; Maciag, T. Post-transcriptional regulation of interleukin 1 alphain various strains of young and senescent human umbilical vein endothelialcells. Proc. Natl. Acad. Sci. USA 1994, 91, 1559–1563. [Google Scholar] [CrossRef] [Green Version]
- He, S.; Sharpless, N.E. Senescence in Health and Disease. Cell 2017, 169, 1000–1011. [Google Scholar] [CrossRef] [Green Version]
- Coppé, J.-P.; Desprez, P.-Y.; Krtolica, A.; Campisi, J. The Senescence-Associated Secretory Phenotype: The Dark Side of Tumor Suppression. Annu. Rev. Pathol. Mech. Dis. 2010, 5, 99–118. [Google Scholar] [CrossRef]
- Takasugi, M.; Okada, R.; Takahashi, A.; Virya Chen, D.; Watanabe, S.; Hara, E. Small extracellular vesicles secreted from senescent cells promote cancer cell proliferation through EphA2. Nat. Commun. 2017, 8, 15729. [Google Scholar] [CrossRef] [Green Version]
- Lehmann, B.D.; Paine, M.S.; Brooks, A.M.; McCubrey, J.A.; Renegar, R.H.; Wang, R.; Terrian, D.M. Senescence-Associated Exosome Release from Human Prostate Cancer Cells. Cancer Res. 2008, 68, 7864–7871. [Google Scholar] [CrossRef] [Green Version]
- Hernandez-Segura, A.; de Jong, T.V.; Melov, S.; Guryev, V.; Campisi, J.; Demaria, M. Unmasking Transcriptional Heterogeneity in Senescent Cells. Curr. Biol. 2017, 27, 2652–2660.e4. [Google Scholar] [CrossRef] [Green Version]
- Kuilman, T.; Michaloglou, C.; Vredeveld, L.C.W.; Douma, S.; van Doorn, R.; Desmet, C.J.; Aarden, L.A.; Mooi, W.J.; Peeper, D.S. Oncogene-Induced Senescence Relayed by an Interleukin-Dependent Inflammatory Network. Cell 2008, 133, 1019–1031. [Google Scholar] [CrossRef] [Green Version]
- Acosta, J.C.; O’Loghlen, A.; Banito, A.; Guijarro, M.V.; Augert, A.; Raguz, S.; Fumagalli, M.; Da Costa, M.; Brown, C.; Popov, N.; et al. Chemokine Signaling via the CXCR2 Receptor Reinforces Senescence. Cell 2008, 133, 1006–1018. [Google Scholar] [CrossRef] [Green Version]
- Orjalo, A.V.; Bhaumik, D.; Gengler, B.K.; Scott, G.K.; Campisi, J. Cell surface-bound IL-1α is an upstream regulator of the senescence-associated IL-6/IL-8 cytokine network. Proc. Natl. Acad. Sci. USA 2009, 106, 17031–17036. [Google Scholar] [CrossRef] [Green Version]
- Bhaumik, D.; Scott, G.K.; Schokrpur, S.; Patil, C.K.; Orjalo, A.V.; Rodier, F.; Lithgow, G.J.; Campisi, J. MicroRNAs miR-146a/b negatively modulate the senescence-associated inflammatory mediators IL-6 and IL-8. Aging 2009, 1, 402–411. [Google Scholar] [CrossRef]
- Chambers, C.R.; Ritchie, S.; Pereira, B.A.; Timpson, P. Overcoming the senescence-associated secretory phenotype (SASP): A complex mechanism of resistance in the treatment of cancer. Mol. Oncol. 2021, 15, 3242–3255. [Google Scholar] [CrossRef]
- Veenstra, J.P.; Bittencourt, L.F.F.; Aird, K.M. The Senescence-Associated Secretory Phenotype in Ovarian Cancer Dissemination. Am. J. Physiol. Physiol. 2022, 323, C125–C132. [Google Scholar] [CrossRef]
- Di, G.; Liu, Y.; Lu, Y.; Liu, J.; Wu, C.; Duan, H.-F. IL-6 Secreted from Senescent Mesenchymal Stem Cells Promotes Proliferation and Migration of Breast Cancer Cells. PLoS ONE 2014, 9, e113572. [Google Scholar] [CrossRef] [PubMed]
- Miao, J.-W.; Liu, L.-J.; Huang, J. Interleukin-6-induced epithelial-mesenchymal transition through signal transducer and activator of transcription 3 in human cervical carcinoma. Int. J. Oncol. 2014, 45, 165–176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Goulet, C.R.; Champagne, A.; Bernard, G.; Vandal, D.; Chabaud, S.; Pouliot, F.; Bolduc, S. Cancer-associated fibroblasts induce epithelial–mesenchymal transition of bladder cancer cells through paracrine IL-6 signalling. BMC Cancer 2019, 19, 137. [Google Scholar] [CrossRef] [Green Version]
- Wang, L.; Tang, C.; Cao, H.; Li, K.; Pang, X.; Zhong, L.; Dang, W.; Tang, H.; Huang, Y.; Wei, L.; et al. Activation of IL-8 via PI3K/Akt-dependent pathway is involved in leptin-mediated epithelial-mesenchymal transition in human breast cancer cells. Cancer Biol. Ther. 2015, 16, 1220–1230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fisher, D.T.; Appenheimer, M.M.; Evans, S.S. The two faces of IL-6 in the tumor microenvironment. Semin. Immunol. 2014, 26, 38–47. [Google Scholar] [CrossRef] [Green Version]
- Waugh, D.J.J.; Wilson, C. The Interleukin-8 Pathway in Cancer. Clin. Cancer Res. 2008, 14, 6735–6741. [Google Scholar] [CrossRef] [Green Version]
- Tamm, I.; Kikuchi, T.; Cardinale, I.; Krueger, J.G. Cell-adhesion-disrupting action of interleukin 6 in human ductal breast carcinoma cells. Proc. Natl. Acad. Sci. USA 1994, 91, 3329–3333. [Google Scholar] [CrossRef] [Green Version]
- Ancrile, B.; Lim, K.-H.; Counter, C.M. Oncogenic Ras-induced secretion of IL6 is required for tumorigenesis. Genes Dev. 2007, 21, 1714–1719. [Google Scholar] [CrossRef] [Green Version]
- du Plessis, M.; Davis, T.; Loos, B.; Pretorius, E.; de Villiers, W.J.S.; Engelbrecht, A.M. Molecular regulation of autophagy in a pro-inflammatory tumour microenvironment: New insight into the role of serum amyloid A. Cytokine Growth Factor Rev. 2021, 59, 71–83. [Google Scholar] [CrossRef]
- Hagihara, K.; Kagawa, S.; Kishida, Y.; Arimitsu, J. Anti-Cytokine Therapy for AA Amyloidosis. In Amyloidosis; InTech: London, UK, 2013. [Google Scholar]
- Paret, C.; Schön, Z.; Szponar, A.; Kovacs, G. Inflammatory Protein Serum Amyloid A1 Marks a Subset of Conventional Renal Cell Carcinomas with Fatal Outcome. Eur. Urol. 2010, 57, 859–866. [Google Scholar] [CrossRef]
- Abouelasrar Salama, S.; De Bondt, M.; De Buck, M.; Berghmans, N.; Proost, P.; Oliveira, V.L.S.; Amaral, F.A.; Gouwy, M.; Van Damme, J.; Struyf, S. Serum Amyloid A1 (SAA1) Revisited: Restricted Leukocyte-Activating Properties of Homogeneous SAA1. Front. Immunol. 2020, 11, 843. [Google Scholar] [CrossRef]
- Kumra, H.; Reinhardt, D.P. Fibronectin-targeted drug delivery in cancer. Adv. Drug Deliv. Rev. 2016, 97, 101–110. [Google Scholar] [CrossRef]
- Ruggiano, A.; Foresti, O.; Carvalho, P. ER-associated degradation: Protein quality control and beyond. J. Cell Biol. 2014, 204, 869–879. [Google Scholar] [CrossRef] [Green Version]
- Lin, T.-C.; Yang, C.-H.; Cheng, L.-H.; Chang, W.-T.; Lin, Y.-R.; Cheng, H.-C. Fibronectin in Cancer: Friend or Foe. Cells 2019, 9, 27. [Google Scholar] [CrossRef] [Green Version]
- Dimri, G.P.; Lee, X.; Basile, G.; Acosta, M.; Scott, G.; Roskelley, C.; Medrano, E.E.; Linskens, M.; Rubelj, I.; Pereira-Smith, O. A biomarker that identifies senescent human cells in culture and in aging skin in vivo. Proc. Natl. Acad. Sci. USA 1995, 92, 9363–9367. [Google Scholar] [CrossRef] [Green Version]
- Chen, A.; Sceneay, J.; Gödde, N.; Kinwel, T.; Ham, S.; Thompson, E.W.; Humbert, P.O.; Möller, A. Intermittent hypoxia induces a metastatic phenotype in breast cancer. Oncogene 2018, 37, 4214–4225. [Google Scholar] [CrossRef]
- Kaushik, S.; Pickup, M.W.; Weaver, V.M. From transformation to metastasis: Deconstructing the extracellular matrix in breast cancer. Cancer Metastasis Rev. 2016, 35, 655–667. [Google Scholar] [CrossRef]
- Casazza, A.; Di Conza, G.; Wenes, M.; Finisguerra, V.; Deschoemaeker, S.; Mazzone, M. Tumor stroma: A complexity dictated by the hypoxic tumor microenvironment. Oncogene 2014, 33, 1743–1754. [Google Scholar] [CrossRef] [Green Version]
- Laitala, A.; Erler, J.T. Hypoxic Signalling in Tumour Stroma. Front. Oncol. 2018, 8, 189. [Google Scholar] [CrossRef]
- Aceto, N.; Bardia, A.; Miyamoto, D.T.; Donaldson, M.C.; Wittner, B.S.; Spencer, J.A.; Yu, M.; Pely, A.; Engstrom, A.; Zhu, H.; et al. Circulating tumor cell clusters are oligoclonal precursors of breast cancer metastasis. Cell 2014, 158, 1110–1122. [Google Scholar] [CrossRef] [Green Version]
- Cheung, K.J.; Ewald, A.J. A collective route to metastasis: Seeding by tumor cell clusters. Science 2016, 352, 167–169. [Google Scholar] [CrossRef] [PubMed]
- Hong, Y.; Fang, F.; Zhang, Q. Circulating tumor cell clusters: What we know and what we expect (Review). Int. J. Oncol. 2016, 49, 2206–2216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yin, T.; Getsios, S.; Caldelari, R.; Kowalczyk, A.P.; Müller, E.J.; Jones, J.C.R.; Green, K.J. Plakoglobin suppresses keratinocyte motility through both cell–cell adhesion-dependent and -independent mechanisms. Proc. Natl. Acad. Sci. USA 2005, 102, 5420–5425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kitamura, T.; Qian, B.-Z.; Pollard, J.W. Immune cell promotion of metastasis. Nat. Rev. Immunol. 2015, 15, 73–86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Paffenholz, S.V.; Salvagno, C.; Ho, Y.-J.; Limjoco, M.; Baslan, T.; Tian, S.; Kulick, A.; de Stanchina, E.; Wilkinson, J.E.; Barriga, F.M.; et al. Senescence induction dictates response to chemo- and immunotherapy in preclinical models of ovarian cancer. Proc. Natl. Acad. Sci. USA 2022, 119, e2117754119. [Google Scholar] [CrossRef] [PubMed]
- Hao, X.; Zhao, B.; Zhou, W.; Liu, H.; Fukumoto, T.; Gabrilovich, D.; Zhang, R. Sensitization of ovarian tumor to immune checkpoint blockade by boosting senescence-associated secretory phenotype. iScience 2021, 24, 102016. [Google Scholar] [CrossRef]
- Tasdemir, N.; Banito, A.; Roe, J.-S.; Alonso-Curbelo, D.; Camiolo, M.; Tschaharganeh, D.F.; Huang, C.-H.; Aksoy, O.; Bolden, J.E.; Chen, C.-C.; et al. BRD4 Connects Enhancer Remodeling to Senescence Immune Surveillance. Cancer Discov. 2016, 6, 612–629. [Google Scholar] [CrossRef] [Green Version]
- Mavrogonatou, E.; Pratsinis, H.; Kletsas, D. The role of senescence in cancer development. Semin. Cancer Biol. 2020, 62, 182–191. [Google Scholar] [CrossRef]
- Luo, X.; Fu, Y.; Loza, A.J.; Murali, B.; Leahy, K.M.; Ruhland, M.K.; Gang, M.; Su, X.; Zamani, A.; Shi, Y.; et al. Stromal-Initiated Changes in the Bone Promote Metastatic Niche Development. Cell Rep. 2016, 14, 82–92. [Google Scholar] [CrossRef] [Green Version]
- Ruhland, M.K.; Loza, A.J.; Capietto, A.-H.; Luo, X.; Knolhoff, B.L.; Flanagan, K.C.; Belt, B.A.; Alspach, E.; Leahy, K.; Luo, J.; et al. Stromal senescence establishes an immunosuppressive microenvironment that drives tumorigenesis. Nat. Commun. 2016, 7, 11762. [Google Scholar] [CrossRef] [Green Version]
- Ohuchida, K.; Mizumoto, K.; Murakami, M.; Qian, L.-W.; Sato, N.; Nagai, E.; Matsumoto, K.; Nakamura, T.; Tanaka, M. Radiation to Stromal Fibroblasts Increases Invasiveness of Pancreatic Cancer Cells through Tumor-Stromal Interactions. Cancer Res. 2004, 64, 3215–3222. [Google Scholar] [CrossRef]
- Birchmeier, C.; Birchmeier, W.; Gherardi, E.; Vande Woude, G.F. Met, metastasis, motility and more. Nat. Rev. Mol. Cell Biol. 2003, 4, 915–925. [Google Scholar] [CrossRef]
- Orr, F.W.; Wang, H.H. Tumor cell interactions with the microvasculature: A rate-limiting step in metastasis. Surg. Oncol. Clin. N. Am. 2001, 10, 357–381. [Google Scholar] [CrossRef]
- Wang, Y.; Zong, X.; Mitra, S.; Mitra, A.K.; Matei, D.; Nephew, K.P. IL-6 mediates platinum-induced enrichment of ovarian cancer stem cells. JCI Insight 2018, 3, e122360. [Google Scholar] [CrossRef] [Green Version]
- So, K.A.; Min, K.J.; Hong, J.H.; Lee, J.-K. Interleukin-6 expression by interactions between gynecologic cancer cells and human mesenchymal stem cells promotes epithelial-mesenchymal transition. Int. J. Oncol. 2015, 47, 1451–1459. [Google Scholar] [CrossRef] [Green Version]
- Macciò, A.; Madeddu, C. Inflammation and ovarian cancer. Cytokine 2012, 58, 133–147. [Google Scholar] [CrossRef] [Green Version]
- Wen, J.; Zhao, Z.; Huang, L.; Wang, L.; Miao, Y.; Wu, J. IL-8 promotes cell migration through regulating EMT by activating the Wnt/β-catenin pathway in ovarian cancer. J. Cell. Mol. Med. 2020, 24, 1588–1598. [Google Scholar] [CrossRef] [Green Version]
- Ni, C.; Huang, J. Dynamic regulation of cancer stem cells and clinical challenges. Clin. Transl. Oncol. 2013, 15, 253–258. [Google Scholar] [CrossRef]
- Batlle, E.; Clevers, H. Cancer stem cells revisited. Nat. Med. 2017, 23, 1124–1134. [Google Scholar] [CrossRef]
- López de Andrés, J.; Griñán-Lisón, C.; Jiménez, G.; Marchal, J.A. Cancer stem cell secretome in the tumor microenvironment: A key point for an effective personalized cancer treatment. J. Hematol. Oncol. 2020, 13, 136. [Google Scholar] [CrossRef]
- Prieto-Vila, M.; Takahashi, R.; Usuba, W.; Kohama, I.; Ochiya, T. Drug Resistance Driven by Cancer Stem Cells and Their Niche. Int. J. Mol. Sci. 2017, 18, 2574. [Google Scholar] [CrossRef] [PubMed]
- Ekström, E.J.; Bergenfelz, C.; von Bülow, V.; Serifler, F.; Carlemalm, E.; Jönsson, G.; Andersson, T.; Leandersson, K. WNT5A induces release of exosomes containing pro-angiogenic and immunosuppressive factors from malignant melanoma cells. Mol. Cancer 2014, 13, 88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Skog, J.; Würdinger, T.; van Rijn, S.; Meijer, D.H.; Gainche, L.; Curry, W.T.; Carter, B.S.; Krichevsky, A.M.; Breakefield, X.O. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat. Cell Biol. 2008, 10, 1470–1476. [Google Scholar] [CrossRef] [PubMed]
- Taraboletti, G.; D’Ascenzoy, S.; Giusti, I.; Marchetti, D.; Borsotti, P.; Millimaggi, D.; Giavazzi, R.; Pavan, A.; Dolo, V. Bioavailability of VEGF in Tumor-Shed Vesicles Depends on Vesicle Burst Induced by Acidic pH. Neoplasia 2006, 8, 96–103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Webber, J.; Steadman, R.; Mason, M.D.; Tabi, Z.; Clayton, A. Cancer Exosomes Trigger Fibroblast to Myofibroblast Differentiation. Cancer Res. 2010, 70, 9621–9630. [Google Scholar] [CrossRef] [Green Version]
- Kim, J.; Kim, T.Y.; Lee, M.S.; Mun, J.Y.; Ihm, C.; Kim, S.A. Exosome cargo reflects TGF-β1-mediated epithelial-to-mesenchymal transition (EMT) status in A549 human lung adenocarcinoma cells. Biochem. Biophys. Res. Commun. 2016, 478, 643–648. [Google Scholar] [CrossRef]
- Gu, J.; Qian, H.; Shen, L.; Zhang, X.; Zhu, W.; Huang, L.; Yan, Y.; Mao, F.; Zhao, C.; Shi, Y.; et al. Gastric Cancer Exosomes Trigger Differentiation of Umbilical Cord Derived Mesenchymal Stem Cells to Carcinoma-Associated Fibroblasts through TGF-β/Smad Pathway. PLoS ONE 2012, 7, e52465. [Google Scholar] [CrossRef] [Green Version]
- Chen, X.; Liang, H.; Zhang, J.; Zen, K.; Zhang, C.-Y. Secreted microRNAs: A new form of intercellular communication. Trends Cell Biol. 2012, 22, 125–132. [Google Scholar] [CrossRef]
- Zhao, Y.; Dong, Q.; Li, J.; Zhang, K.; Qin, J.; Zhao, J.; Sun, Q.; Wang, Z.; Wartmann, T.; Jauch, K.W.; et al. Targeting cancer stem cells and their niche: Perspectives for future therapeutic targets and strategies. Semin. Cancer Biol. 2018, 53, 139–155. [Google Scholar] [CrossRef]
- Najafi, M.; Mortezaee, K.; Majidpoor, J. Cancer stem cell (CSC) resistance drivers. Life Sci. 2019, 234, 116781. [Google Scholar] [CrossRef]
- Cojoc, M.; Peitzsch, C.; Trautmann, F.; Polishchuk, L.; Telegeev, G.D.; Dubrovska, A. Emerging targets in cancer management: Role of the CXCL12/CXCR4 axis. Onco. Targets. Ther. 2013, 6, 1347–1361. [Google Scholar] [CrossRef]
- Roato, I.; Ferracini, R. Cancer Stem Cells, Bone and Tumor Microenvironment: Key Players in Bone Metastases. Cancers 2018, 10, 56. [Google Scholar] [CrossRef] [Green Version]
- Es-haghi, M.; Soltanian, S.; Dehghani, H. Perspective: Cooperation of Nanog, NF-κΒ, and CXCR4 in a regulatory network for directed migration of cancer stem cells. Tumor Biol. 2016, 37, 1559–1565. [Google Scholar] [CrossRef]
- Zhang, Q.; Han, Z.; Zhu, Y.; Chen, J.; Li, W. Role of hypoxia inducible factor-1 in cancer stem cells (Review). Mol. Med. Rep. 2020, 23, 17. [Google Scholar] [CrossRef]
- Tong, W.-W.; Tong, G.-H.; Liu, Y. Cancer stem cells and hypoxia-inducible factors (Review). Int. J. Oncol. 2018, 53, 469–476. [Google Scholar] [CrossRef] [Green Version]
- Jiménez, G.; López de Andrés, J.; Marchal, J.A. Stem Cell-Secreted Factors in the Tumor Microenvironment. In Tumor Microenvironment; Springer: Cham, Switzerland, 2020; pp. 115–126. [Google Scholar]
- Eales, K.L.; Hollinshead, K.E.R.; Tennant, D.A. Hypoxia and metabolic adaptation of cancer cells. Oncogenesis 2016, 5, e190. [Google Scholar] [CrossRef] [Green Version]
- Jia, D.; Li, L.; Andrew, S.; Allan, D.; Li, X.; Lee, J.; Ji, G.; Yao, Z.; Gadde, S.; Figeys, D.; et al. An autocrine inflammatory forward-feedback loop after chemotherapy withdrawal facilitates the repopulation of drug-resistant breast cancer cells. Cell Death Dis. 2017, 8, e2932. [Google Scholar] [CrossRef] [Green Version]
- Korkaya, H.; Kim, G.; Davis, A.; Malik, F.; Henry, N.L.; Ithimakin, S.; Quraishi, A.A.; Tawakkol, N.; D’Angelo, R.; Paulson, A.K.; et al. Activation of an IL6 Inflammatory Loop Mediates Trastuzumab Resistance in HER2+ Breast Cancer by Expanding the Cancer Stem Cell Population. Mol. Cell 2012, 47, 570–584. [Google Scholar] [CrossRef] [Green Version]
- Bruna, A.; Greenwood, W.; Le Quesne, J.; Teschendorff, A.; Miranda-Saavedra, D.; Rueda, O.M.; Sandoval, J.L.; Vidakovic, A.T.; Saadi, A.; Pharoah, P.; et al. TGFβ induces the formation of tumour-initiating cells in claudinlow breast cancer. Nat. Commun. 2012, 3, 1055. [Google Scholar] [CrossRef]
- Deep, G.; Jain, A.; Kumar, A.; Agarwal, C.; Kim, S.; Leevy, W.M.; Agarwal, R. Exosomes secreted by prostate cancer cells under hypoxia promote matrix metalloproteinases activity at pre-metastatic niches. Mol. Carcinog. 2020, 59, 323–332. [Google Scholar] [CrossRef]
- Fluegen, G.; Avivar-Valderas, A.; Wang, Y.; Padgen, M.R.; Williams, J.K.; Nobre, A.R.; Calvo, V.; Cheung, J.F.; Bravo-Cordero, J.J.; Entenberg, D.; et al. Phenotypic heterogeneity of disseminated tumour cells is preset by primary tumour hypoxic microenvironments. Nat. Cell Biol. 2017, 19, 120–132. [Google Scholar] [CrossRef] [PubMed]
- Ghajar, C.M.; Peinado, H.; Mori, H.; Matei, I.R.; Evason, K.J.; Brazier, H.; Almeida, D.; Koller, A.; Hajjar, K.A.; Stainier, D.Y.R.; et al. The perivascular niche regulates breast tumour dormancy. Nat. Cell Biol. 2013, 15, 807–817. [Google Scholar] [CrossRef] [PubMed]
- Cui, D.; Huang, Z.; Liu, Y.; Ouyang, G. The multifaceted role of periostin in priming the tumor microenvironments for tumor progression. Cell. Mol. Life Sci. 2017, 74, 4287–4291. [Google Scholar] [CrossRef] [PubMed]
- Durrett, R.; Foo, J.; Leder, K.; Mayberry, J.; Michor, F. Intratumor Heterogeneity in Evolutionary Models of Tumor Progression. Genetics 2011, 188, 461–477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aguirre-Ghiso, J.A.; Liu, D.; Mignatti, A.; Kovalski, K.; Ossowski, L. Urokinase Receptor and Fibronectin Regulate the ERK MAPK to p38 MAPK Activity Ratios That Determine Carcinoma Cell Proliferation or Dormancy In Vivo. Mol. Biol. Cell 2001, 12, 863–879. [Google Scholar] [CrossRef] [Green Version]
- Barkan, D.; El Touny, L.H.; Michalowski, A.M.; Smith, J.A.; Chu, I.; Davis, A.S.; Webster, J.D.; Hoover, S.; Simpson, R.M.; Gauldie, J.; et al. Metastatic Growth from Dormant Cells Induced by a Col-I–Enriched Fibrotic Environment. Cancer Res. 2010, 70, 5706–5716. [Google Scholar] [CrossRef] [Green Version]
- Pollard, J.W. Tumour-educated macrophages promote tumour progression and metastasis. Nat. Rev. Cancer 2004, 4, 71–78. [Google Scholar] [CrossRef]
- Li, Z.; Liu, F.; Kirkwood, K.L. The p38/MKP-1 signaling axis in oral cancer: Impact of tumor-associated macrophages. Oral Oncol. 2020, 103, 104591. [Google Scholar] [CrossRef]
- Kale, S.; Raja, R.; Thorat, D.; Soundararajan, G.; Patil, T.V.; Kundu, G.C. Osteopontin signaling upregulates cyclooxygenase-2 expression in tumor-associated macrophages leading to enhanced angiogenesis and melanoma growth via α9β1 integrin. Oncogene 2014, 33, 2295–2306. [Google Scholar] [CrossRef]
- Jones, S.A.; Jenkins, B.J. Recent insights into targeting the IL-6 cytokine family in inflammatory diseases and cancer. Nat. Rev. Immunol. 2018, 18, 773–789. [Google Scholar] [CrossRef]
- Wang, L.; Cao, L.; Wang, H.; Liu, B.; Zhang, Q.; Meng, Z.; Wu, X.; Zhou, Q.; Xu, K. Cancer-associated fibroblasts enhance metastatic potential of lung cancer cells through IL-6/STAT3 signaling pathway. Oncotarget 2017, 8, 76116–76128. [Google Scholar] [CrossRef]
- Radharani, N.N.V.; Yadav, A.S.; Nimma, R.; Kumar, T.V.S.; Bulbule, A.; Chanukuppa, V.; Kumar, D.; Patnaik, S.; Rapole, S.; Kundu, G.C. Tumor-associated macrophage derived IL-6 enriches cancer stem cell population and promotes breast tumor progression via Stat-3 pathway. Cancer Cell Int. 2022, 22, 122. [Google Scholar] [CrossRef]
- Nagasaki, T.; Hara, M.; Nakanishi, H.; Takahashi, H.; Sato, M.; Takeyama, H. Interleukin-6 released by colon cancer-associated fibroblasts is critical for tumour angiogenesis: Anti-interleukin-6 receptor antibody suppressed angiogenesis and inhibited tumour–stroma interaction. Br. J. Cancer 2014, 110, 469–478. [Google Scholar] [CrossRef] [Green Version]
- Wei, L.-H.; Kuo, M.-L.; Chen, C.-A.; Chou, C.-H.; Lai, K.-B.; Lee, C.-N.; Hsieh, C.-Y. Interleukin-6 promotes cervical tumor growth by VEGF-dependent angiogenesis via a STAT3 pathway. Oncogene 2003, 22, 1517–1527. [Google Scholar] [CrossRef] [Green Version]
- Butti, R.; Nimma, R.; Kundu, G.; Bulbule, A.; Kumar, T.V.S.; Gunasekaran, V.P.; Tomar, D.; Kumar, D.; Mane, A.; Gill, S.S.; et al. Tumor-derived osteopontin drives the resident fibroblast to myofibroblast differentiation through Twist1 to promote breast cancer progression. Oncogene 2021, 40, 2002–2017. [Google Scholar] [CrossRef]
Molecule | Effect on Premetastatic Niche Preparation | ||
---|---|---|---|
ECM (Extracellular Matrix) Changes | Cell Recruitment and Activity | Vascularization | |
LOX | Collagen crosslinking ↑Matrix stiffness ↑Fibronectin ↑Osteolysis | CD11b+ BMDCs and MDSCs recruitment ↑Proliferative and invasive phenotype | ↑Angiogenesis |
VEGF | ↑Fibronectin ↑Osteolysis and bone reabsortion ↑Bone sialoprotein production MMP-9 secretion | CD11b+ BMDCs and VEGFR+ BMDCs recruitment CXCR2+ tumor cells recruitment Nesting induction ↑Adhesion ↑Invadipodia formation | ↑Angiogenesis ↑Permeabilization |
TGFb | Fibronectin, collagen and periostin production ↑Bone lesions | MDSCs recruitment Mesothelial to mesenchymal transition Immune suppression of NKs, d T cells and CD8+ T cells N2 and M2 polarization CCL9 expression in CD11b+ BMDCs | / |
CAIX | ↑Intregin expression ↑MMP-14 expression ↑MMP-9 secretion | ↑Cell motility IFNg production suppression G-CSF production Inhibits NK citotoxicity | / |
S100A8 and S100A9 | MMP-9 secretion Versican deposit | CD11b+ BMDCs recruitment Invasive phenotype Increased invadipodia formation Chronic inflammation | / |
SAA3 | Mac1+ recruitment Circulating tumor cells recruitment ↑ TNFa production Chronic inflammation | / | |
IL-6 | Osteolysis and bone reabsorption MMP-9 secretion | MDSCs recruitment CD11b+ osteoclastic differentiation Immunosuppresive niche | CCL5 expression in LECs |
Global effect | Creates physical space Releases trapped molecules Increases cell adhesion and nesting Difficult access to eliminate tumor cells | Immune suppressed secondary site Increased tumor cell arrival | Favors circulating CSCs arrival Nutrient arrival to micrometastasis |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Clemente-González, C.; Carnero, A. Role of the Hypoxic-Secretome in Seed and Soil Metastatic Preparation. Cancers 2022, 14, 5930. https://doi.org/10.3390/cancers14235930
Clemente-González C, Carnero A. Role of the Hypoxic-Secretome in Seed and Soil Metastatic Preparation. Cancers. 2022; 14(23):5930. https://doi.org/10.3390/cancers14235930
Chicago/Turabian StyleClemente-González, Cynthia, and Amancio Carnero. 2022. "Role of the Hypoxic-Secretome in Seed and Soil Metastatic Preparation" Cancers 14, no. 23: 5930. https://doi.org/10.3390/cancers14235930