Skip to main content

In vivo imaging of inflammatory response in cancer research

Abstract

Inflammation can contribute to the development and progression of cancer. The inflammatory responses in the tumor microenvironment are shaped by complex sequences of dynamic intercellular cross-talks among diverse types of cells, and recapitulation of these dynamic events in vitro has yet to be achieved. Today, intravital microscopy with two-photon excitation microscopes (2P-IVM) is the mainstay technique for observing intercellular cross-talks in situ, unraveling cellular and molecular mechanisms in the context of their spatiotemporal dynamics. In this review, we summarize the current state of 2P-IVM with fluorescent indicators of signal transduction to reveal the cross-talks between cancer cells and surrounding cells including both immune and non-immune cells. We also discuss the potential application of red-shifted indicators along with optogenetic tools to 2P-IVM. In an era of single-cell transcriptomics and data-driven research, 2P-IVM will remain a key advantage in delivering the missing spatiotemporal context in the field of cancer research.

Background

Inflammation is part of the complex biological response triggered by the immune response to infection, injury, and other environmental challenges. Inflammation functions not only as a host defense process, but also as a tissue repair, regeneration, and remodeling process, modulating tissue homeostasis [1]. During the last couple of decades, it is increasingly appreciated that inflammation is an important component of tumorigenesis ranging from cancer development, progression, and therapy resistance [2]. Large epidemiological clinical studies clarifying the effects of non-steroidal anti-inflammatory drugs (NSAIDs) [3, 4] or interleukin 1β inhibitors [5] on reducing incidence and mortality in many cancer types have provided excellent evidence of the supportive role of inflammation in tumorigenesis. Defining the molecular and cellular cross-talks underlying tumor-promoting inflammation will be essential for further development of anti-cancer therapies.

Inflammatory responses are shaped by dynamic sequential inter-cellular cross-talks between a multitude of functionally diverse types of cells of innate and adaptive immunity. The development of high-dimensional profiling technologies such as cytometry by time of flight (CyTOF) and single-cell RNA sequencing (scRNA-seq) has enabled to profile of a vast heterogeneity of immune cell states and functionality. However, how different cellular components engage in the sequential inflammatory responses, and how these events take place in a spatially organized manner within the tumor microenvironment remain unsolved questions. Despite the recent advancement of organoid culture systems for modeling the tumor microenvironment [6, 7], recapitulation of inflammatory responses in vitro has yet to be achieved. A comprehensive understanding of this continuously dynamic and complex process requires in situ investigation in living organisms.

Intravital microscopy with two-photon excitation microscopy (2P-IVM)

Intravital microscopy (IVM) is a unique imaging method that allows observing biological processes in living organisms. The invention of the microscope leads the early pioneers to observe living cells in the seventeenth century [8]. Despite this long history, it was only after the widespread application of a two-photon excitation microscope (2PEM) that the number of studies using IVM started to increase [9, 10]. Two-photon excitation arises from the simultaneous absorption of two photons in a single event, which depends on the square of the light intensity. Due to this property, the probability of two-photon absorption at the center of focus is substantially greater than outside of the focus, allowing little excitation of fluorophores along the optical path in contrast to the conventional single-photon absorption (Fig. 1) [11]. This localization of two-photon excitation to solely the focal plane provides several advantages over conventional microscopy, including the maximum recovery of photons from the molecules of interest on focal planes, the reduction of photodamage throughout the sample, and the restricted out-of-focus absorption allowing deep tissue penetration [12]. With these important principles, 2PEM now allows us to observe brain tissue to more than 1-mm depth, in contrast to conventional microscopy, which can visualize only structures close to the surface of tissues [13].

Fig. 1
figure 1

Properties of two-photon excitation microscopy (2PEM). In conventional single-photon fluorescence microscopy, fluorescence molecules along the light path are excited. In 2PEM, two photons must reach the single fluorophore almost simultaneously, which occurs only at the focal plane, markedly reducing the background fluorescence

Initially, after being introduced into the field of immunology, 2P-IVM provided significant insights into the dynamic cellular interplay between different fluorescently labeled cell populations [14,15,16]. Using pre-clinical tumor-bearing mouse models, 2P-IVM has revealed the inter-cellular cross-talks within the tumor microenvironment in numerous anatomical sites, including the skin [17, 18], lungs [19, 20], digestive tract [21], lymph nodes [22], bone marrow [23], and brain [24,25,26]. Also, the application of autofluorescence imaging along with fluorescent labeling that measures functional parameters like NAD(P)H and FAD for monitoring metabolic activity has provided an additional layer of information on immune cells [27].

Then, along with the development and application of fluorescent indicators, live signal transduction events now can be observed as cells actively engage in interactions or receive soluble stimuli [28]. Below, we will consider some applications of 2P-IVM with fluorescent indicators of signal transduction in the field of cancer research in detail.

Visualization of PGE2 secretion from tumor cells

Prostaglandin E2 (PGE2) is the most widely produced prostanoid in the body, which can modulate various steps of inflammation in both pro-inflammatory and anti-inflammatory manners [29]. PGE2 is known to promote tumor development by several mechanisms that promote cell growth, invasion, migration, angiogenesis, and immune evasion [30,31,32]. Currently, it is believed that the concentration of PGE2 within tumor tissues is regulated mostly by transcriptional regulation of Ptgs2 encoding cyclooxygenase-2 (COX-2) which is the rate-limiting enzyme in the PGE2 synthesis pathway [33]. However, the question remains of how the PGE2 secretion from tumor cells is regulated in the tumor microenvironment in the context of cellular cross-talks.

To monitor PGE2 secretion using 2P-IVM, we focused on Ca2+ signal transduction in tumor cells. The Ca2+-induced activation of phospholipase A1 (PLA2), particularly cytosolic phospholipase A2 (cPLA2a), is recognized as the rate-limiting step in PGE2 secretion, which liberates arachidonic acid from cell membrane phospholipids [34, 35]. Therefore, we hypothesized that the Ca2+ transients in tumor cells can work as a surrogate marker for PGE2 secretion [36]. The Ca2+ transients in mouse melanoma cells in the tumor microenvironment were visualized by using a genetically encoded calcium indicator, GCaMP6s (Fig. 2A) [37]. Of note, the Ca2+ transients observed in tumor cells were significantly suppressed by CRISPR/Cas9-mediated gene knockout of Gnaq (Gnaq−/−), which encodes guanine nucleotide-binding protein G(q) subunit alpha. Importantly, this suppression in Ca2+ transients resulted in a marked reduction of the PGE2 concentration in the tumor microenvironment, supporting the notion that (i) Ca2+ transients reflect PGE2 secretion and (ii) the GqPCR signaling pathway is required for PGE2 secretion from tumor cells in this melanoma model. The downstream analysis revealed that the major GqPCR ligand in this model was thromboxane A2 (TXA2) released from endothelial cells. As shown here, 2P-IVM with the fluorescent indicator of Ca2+ signal transduction enabled us to clarify the indirect intercellular cross-talks between tumor cells and endothelial cells in enhancing PGE2 secretion from tumor cells and thereby promoting tumor immune evasion within the tumor microenvironment.

Fig. 2
figure 2

2P-IVM with fluorescent indicators to reveal the cross-talks between tumor cells and surrounding cells in the tumor microenvironment including both immune and non-immune cells. Indirect cross-talks between A tumor cells and non-immune cells, B tumor cells and immune cells, and C direct cross-talks between tumor cells and immune cells can be visualized by using 2P-IVM with fluorescent indicators (gray cells: tumor cells; magenta cells: tumor cells receiving signals via signaling molecules, TXA2 or IFN-γ; yellow cells: T cells or NK cells). Melanoma cells expressing GCaMP6s were imaged using 2P-IVM (A, bottom). The image represents the maximum intensity projection for 10 min. Images were adapted courtesy of Konishi et al. [36]. Schematic representation of the fluorescent indicator for IFN-γ (B, bottom). Melanoma cells expressing this fluorescent indicator were imaged using 2P-IVM. Images represent the mCherry and turquoise-GL-NLS of parental cells. Images were adapted by courtesy of Tanaka et al. [38]. Melanoma cells expressing GCaMP6s were imaged in the lung using 2P-IVM (C, bottom). Top, FRET/CFP ratio of an NK cell is shown. Melanoma cell is shown in white. GCaMP6s intensity is displayed in pseudo-color (bottom). The NK cell is shown in white. The images were adapted by courtesy of Ichise et al. [39]

Visualization of IFNγ-induced signaling in tumor cells

Using the same mouse melanoma model with 2PM-IVM, Tanaka et al. successfully visualized the tumor cells receiving interferon-gamma (IFN-γ) stimuli in the tumor microenvironment [38]. IFN–γ is a cytokine secreted by immune cells, especially NK cells and T cells, and enhances anti-tumor immune response [40, 41]. Tanaka et al. utilized the dual promoter system with interferon γ-activated sequence (GAS) [42] and developed a fluorescent indicator for IFN-γ-mediated signal transduction in tumor cells (Fig. 2B). This indicator enabled the visualization of tumor cells receiving IFN-γ stimuli as the expression of fluorescent protein at a single-cell resolution. As anticipated, the GAS activity in the Gnaq−/− tumor was significantly higher than that in the parental tumors, supporting the activated anti-tumor immune response with an abundance of IFN-γ. Similarly, Hoekstra et al. analyzed the spatial spreading of T cell-derived IFN-γ in vivo by using an IFN-γ-sensing fluorescent indicator that induces the expression of fluorescent protein after IFN-γ receptor triggering [42]. In addition to these two examples, the spatiotemporal spread of IFN-γ within the tumor microenvironment was also visualized by a fluorescent indicator where the translocation of a fluorescent protein from the cytoplasm to the nucleus represents the receiving of IFN-γ stimuli [43]. These prominent examples support the utility of 2P-IVM in recording indirect cellular cross-talks via soluble stimuli such as IFN-γ.

Visualization of tumor killing by NK cells

To develop optimal immunotherapies, it is important to understand how cytotoxic lymphocytes such as NK cells react to tumor cell challenges and how tumor cells in turn respond. Using 2P-IVM with a metastatic melanoma mouse model, Ichise et al. visualized the direct intercellular cross-talks between NK cells and metastatic tumor cells in the lung (Fig. 2C) [39]. To track NK cell activation, extracellular signal-regulated kinase (ERK) activity was monitored by using a Förester resonance energy transfer (FRET)-based biosensor (EKAREV) [44, 45]. In the meantime, Ca2+ influx in tumor cells was monitored by using GCaMP6s [46] as a surrogate marker for apoptosis. Using this system, they revealed that ERK activation in NK cells contributes to tumor-cell killing, granting further study for underlying mechanisms controlling ERK activity dynamics in NK cells to develop optimal immunotherapies using NK cells. Interestingly, they also reported a marked decrease in the proportion of ERK activation and Ca2+ influx 24 h after tumor injection, suggesting exhaustion of NK cells.

These examples indicate the versatile utility of 2P-IVM with a fluorescent indicator of signal transduction in analyzing inflammatory responses in the tumor microenvironment, encouraging the application of 2P-IVM in the field of cancer research.

Future perspective

Recent advances in optogenetics have opened new routes to cancer research with 2P-IVM. The term “optogenetics” was first coined by Deisseroth et al. in 2006 [47]. Optogenetics is a biological technique that employs natural and engineered photoreceptors to be genetically introduced into the cells of interest, allowing these target cells to be photosensitive and addressable by illumination [48,49,50]. On the level of individual cells, this allows the precise control of cell signaling pathways in time and space. Starting with Channelrhodopsin (ChR) to excite neurons as a prototypical optogenetic tool [51, 52], several optogenetic tools are now available to control the activity of neurons or other cell types with light by regulating protein heterodimerization [53,54,55], homo-dimerization [56, 57], gene expression [58,59,60], protein degradation [61], nuclear-cytosolic protein translocation [62,63,64,65], and liquid–liquid protein phase separation [66,67,68]. Optogenetic manipulation can be achieved by two-photon excitation and is compatible with 2P-IVM. For example, the optogenetic control of protein dimerization to induce signal transduction and gene expression has been achieved by two-photon illumination [69,70,71]. Indeed, Kinjo et al. achieved the simultaneous photoactivation and recording of ERK signaling pathway activity in the mouse epidermal cells at a single-cell resolution during 2P-IVM by introducing both a fluorescent indicator and an optogenetic tool. The recent development of red, near-infrared, and far-red shifted fluorescent indicators including calcium [72, 73], voltage [74, 75], and kinase activity [76] has strengthened the potential for accomplishing 2P-IVM in combination with optogenetic control. The application of red-shifted indicators along with optogenetic tools to 2P-IVM will advance the utility of this versatile tool in the field of cancer research.

Conclusions

In the field of cancer research, major challenges remain in elucidating the dynamic sequential intercellular cross-talks within the tumor microenvironment. The 2P-IVM with fluorescent indicators of signal transduction enables us to investigate this continuously dynamic and complex process in living organisms in situ and real-time. The application of red-shifted indicators along with optogenetic tools will further facilitate the potential of 2P-IVM, revealing unexpected cellular and molecular mechanisms operating in the tumor microenvironment.

Availability of data and materials

Not applicable.

Abbreviations

2P-IVM:

Intravital microscopy with two-photon excitation microscopes

NSAIDs:

Non-steroidal anti-inflammatory drugs

CyTOF:

Cytometry by the time of flight

scRNA-seq:

Single-cell RNA sequencing

2PEM:

Two-photon excitation microscope

PGE2 :

Prostaglandin E2

PLA2:

Phospholipase A1

cPLA2a:

Cytosolic phospholipase A2

TXA2 :

Thromboxane A2

IFN–γ:

Interferon-gamma

ERK:

Extracellular signal-regulated kinase

FRET:

Förester resonance energy transfer

ChR:

Channelrhodopsin

References

  1. Medzhitov R. Origin and physiological roles of inflammation. Nature. 2008;454(7203):428–35.

    Article  CAS  Google Scholar 

  2. Greten FR, Grivennikov SI. Inflammation and cancer: triggers, mechanisms, and consequences. Immunity. 2019;51(1):27–41.

    Article  CAS  Google Scholar 

  3. Rothwell PM, Fowkes FGR, Belch JFF, Ogawa H, Warlow CP, Meade TW. Effect of daily aspirin on long-term risk of death due to cancer: analysis of individual patient data from randomised trials. Lancet. 2011;377(9759):31–41.

    Article  CAS  Google Scholar 

  4. Rothwell PM, Wilson M, Price JF, Belch JFF, Meade TW, Mehta Z. Effect of daily aspirin on risk of cancer metastasis: a study of incident cancers during randomised controlled trials. Lancet. 2012;379(9826):1591–601.

    Article  CAS  Google Scholar 

  5. Ridker PM, MacFadyen JG, Thuren T, Everett BM, Libby P, Glynn RJ, et al. Effect of interleukin-1β inhibition with canakinumab on incident lung cancer in patients with atherosclerosis: exploratory results from a randomised, double-blind, placebo-controlled trial. Lancet. 2017;390(10105):1833–42.

    Article  CAS  Google Scholar 

  6. Jenkins RW, Aref AR, Lizotte PH, Ivanova E, Stinson S, Zhou CW, et al. Ex vivo profiling of PD-1 blockade using organotypic tumor spheroids. Cancer Discov. 2018;8(2):196–215.

    Article  CAS  Google Scholar 

  7. Neal JT, Li X, Zhu J, Giangarra V, Grzeskowiak CL, Ju J, et al. Organoid modeling of the tumor immune microenvironment. Cell. 2018;175(7):1972-88.e16.

    Article  CAS  Google Scholar 

  8. Norman K. Techniques: intravital microscopy–a method for investigating disseminated intravascular coagulation? Trends Pharmacol Sci. 2005;26(6):327–32.

    Article  CAS  Google Scholar 

  9. Secklehner J, Lo Celso C, Carlin LM. Intravital microscopy in historic and contemporary immunology. Immunol Cell Biol. 2017;95(6):506–13.

    Article  Google Scholar 

  10. Matsuda M, Terai K. Experimental pathology by intravital microscopy and genetically encoded fluorescent biosensors. Pathol Int. 2020;70(7):379–90.

    Article  CAS  Google Scholar 

  11. Denk W, Strickler JH, Webb WW. Two-photon laser scanning fluorescence microscopy. Science. 1990;248(4951):73–6.

    Article  CAS  Google Scholar 

  12. Benninger RK, Piston DW. Two-photon excitation microscopy for the study of living cells and tissues. Curr Protoc Cell Biol. 2013;Chapter 4:Unit 4.11.1–24.

  13. Helmchen F, Denk W. Deep tissue two-photon microscopy. Nat Methods. 2005;2(12):932–40.

    Article  CAS  Google Scholar 

  14. Breart B, Lemaître F, Celli S, Bousso P. Two-photon imaging of intratumoral CD8+ T cell cytotoxic activity during adoptive T cell therapy in mice. J Clin Invest. 2008;118(4):1390–7.

    Article  CAS  Google Scholar 

  15. Germain RN, Robey EA, Cahalan MD. A decade of imaging cellular motility and interaction dynamics in the immune system. Science. 2012;336(6089):1676–81.

    Article  CAS  Google Scholar 

  16. Liarski VM, Sibley A, van Panhuys N, Ai J, Chang A, Kennedy D, et al. Quantifying in situ adaptive immune cell cognate interactions in humans. Nat Immunol. 2019;20(4):503–13.

    Article  CAS  Google Scholar 

  17. Boissonnas A, Fetler L, Zeelenberg IS, Hugues S, Amigorena S. In vivo imaging of cytotoxic T cell infiltration and elimination of a solid tumor. J Exp Med. 2007;204(2):345–56.

    Article  CAS  Google Scholar 

  18. Budhu S, Schaer DA, Li Y, Toledo-Crow R, Panageas K, Yang X, et al. Blockade of surface-bound TGF-β on regulatory T cells abrogates suppression of effector T cell function in the tumor microenvironment. Sci Signal. 2017;10(494).

  19. Headley MB, Bins A, Nip A, Roberts EW, Looney MR, Gerard A, et al. Visualization of immediate immune responses to pioneer metastatic cells in the lung. Nature. 2016;531(7595):513–7.

    Article  CAS  Google Scholar 

  20. Hanna RN, Chodaczek G, Hedrick CC. In vivo imaging of tumor and immune cell interactions in the lung. Bio Protoc. 2016;6(20):e1973.

  21. Kagawa Y, Matsumoto S, Kamioka Y, Mimori K, Naito Y, Ishii T, et al. Cell cycle-dependent Rho GTPase activity dynamically regulates cancer cell motility and invasion in vivo. PLoS One. 2013;8(12):e83629.

    Article  Google Scholar 

  22. Boissonnas A, Scholer-Dahirel A, Simon-Blancal V, Pace L, Valet F, Kissenpfennig A, et al. Foxp3<sup>+</sup> T cells induce perforin-dependent dendritic cell death in tumor-draining lymph nodes. Immunity. 2010;32(2):266–78.

    Article  CAS  Google Scholar 

  23. Morimatsu M, Yamashita E, Seno S, Sudo T, Kikuta J, Mizuno H, et al. Migration arrest of chemoresistant leukemia cells mediated by MRTF-SRF pathway. Inflammation and Regeneration. 2020;40(1):15.

    Article  CAS  Google Scholar 

  24. Zomer A, Croci D, Kowal J, van Gurp L, Joyce JA. Multimodal imaging of the dynamic brain tumor microenvironment during glioblastoma progression and in response to treatment. iScience. 2022;25(7):104570.

  25. Zhang W, Karschnia P, von Mücke-Heim IA, Mulazzani M, Zhou X, Blobner J, et al. In vivo two-photon characterization of tumor-associated macrophages and microglia (TAM/M) and CX3CR1 during different steps of brain metastasis formation from lung cancer. Neoplasia. 2021;23(11):1089–100.

    Article  CAS  Google Scholar 

  26. Ricard C, Tchoghandjian A, Luche H, Grenot P, Figarella-Branger D, Rougon G, et al. Phenotypic dynamics of microglial and monocyte-derived cells in glioblastoma-bearing mice. Sci Rep. 2016;6(1):26381.

    Article  CAS  Google Scholar 

  27. Heaster TM, Heaton AR, Sondel PM, Skala MC. Intravital metabolic autofluorescence imaging captures macrophage heterogeneity across normal and cancerous tissue. Front Bioeng Biotechnol. 2021;9:644648.

  28. Hor JL, Germain RN. Intravital and high-content multiplex imaging of the immune system. Trends Cell Biol. 2022;32(5):406–20.

    Article  Google Scholar 

  29. Ricciotti E, FitzGerald GA. Prostaglandins and inflammation. Arterioscler Thromb Vasc Biol. 2011;31(5):986–1000.

    Article  CAS  Google Scholar 

  30. Dannenberg AJ, Subbaramaiah K. Targeting cyclooxygenase-2 in human neoplasia: rationale and promise. Cancer Cell. 2003;4(6):431–6.

    Article  CAS  Google Scholar 

  31. Wang D, Dubois RN. Eicosanoids and cancer. Nat Rev Cancer. 2010;10(3):181–93.

    Article  CAS  Google Scholar 

  32. Kalinski P. Regulation of immune responses by prostaglandin E2. J Immunol. 2012;188(1):21–8.

    Article  CAS  Google Scholar 

  33. Greenhough A, Smartt HJ, Moore AE, Roberts HR, Williams AC, Paraskeva C, et al. The COX-2/PGE2 pathway: key roles in the hallmarks of cancer and adaptation to the tumour microenvironment. Carcinogenesis. 2009;30(3):377–86.

    Article  CAS  Google Scholar 

  34. Kita Y, Shindou H, Shimizu T. Cytosolic phospholipase A(2) and lysophospholipid acyltransferases. Biochim Biophys Acta Mol Cell Biol Lipids. 2019;1864(6):838–45.

    Article  CAS  Google Scholar 

  35. Park JY, Pillinger MH, Abramson SB. Prostaglandin E2 synthesis and secretion: the role of PGE2 synthases. Clin Immunol. 2006;119(3):229–40.

    Article  CAS  Google Scholar 

  36. Konishi Y, Ichise H, Watabe T, Oki C, Tsukiji S, Hamazaki Y, et al. Intravital imaging identifies the VEGF–TXA2 axis as a critical promoter of PGE2 secretion from tumor cells and immune evasion. Can Res. 2021;81(15):4124–32.

    Article  CAS  Google Scholar 

  37. Chen T-W, Wardill TJ, Sun Y, Pulver SR, Renninger SL, Baohan A, et al. Ultrasensitive fluorescent proteins for imaging neuronal activity. Nature. 2013;499(7458):295–300.

    Article  CAS  Google Scholar 

  38. Tanaka T, Konishi Y, Ichise H, Tsukiji S, Matsuda M, Terai K. A dual promoter system to monitor IFN-γ signaling in vivo at single-cell resolution. Cell Struct Funct. 2021;46(2):103–11.

    Article  CAS  Google Scholar 

  39. Ichise H, Tsukamoto S, Hirashima T, Konishi Y, Oki C, Tsukiji S, et al. Functional visualization of NK cell-mediated killing of metastatic single tumor cells. eLife. 2022;11:e76269.

  40. Kaplan DH, Shankaran V, Dighe AS, Stockert E, Aguet M, Old LJ, et al. Demonstration of an interferon gamma-dependent tumor surveillance system in immunocompetent mice. Proc Natl Acad Sci U S A. 1998;95(13):7556–61.

    Article  CAS  Google Scholar 

  41. Dighe AS, Richards E, Old LJ, Schreiber RD. Enhanced in vivo growth and resistance to rejection of tumor cells expressing dominant negative IFN gamma receptors. Immunity. 1994;1(6):447–56.

    Article  CAS  Google Scholar 

  42. Hoekstra ME, Bornes L, Dijkgraaf FE, Philips D, Pardieck IN, Toebes M, et al. Long-distance modulation of bystander tumor cells by CD8(+) T cell-secreted IFNγ. Nat Cancer. 2020;1(3):291–301.

    Article  CAS  Google Scholar 

  43. Thibaut R, Bost P, Milo I, Cazaux M, Lemaître F, Garcia Z, et al. Bystander IFN-γ activity promotes widespread and sustained cytokine signaling altering the tumor microenvironment. Nature Cancer. 2020;1(3):302–14.

    Article  CAS  Google Scholar 

  44. Konishi Y, Terai K, Furuta Y, Kiyonari H, Abe T, Ueda Y, et al. Live-cell FRET imaging reveals a role of extracellular signal-regulated kinase activity dynamics in thymocyte motility. iScience. 2018;10:98–113.

  45. Komatsu N, Terai K, Imanishi A, Kamioka Y, Sumiyama K, Jin T, et al. A platform of BRET-FRET hybrid biosensors for optogenetics, chemical screening, and in vivo imaging. Sci Rep. 2018;8(1):8984.

    Article  Google Scholar 

  46. Bosschaart N, Edelman GJ, Aalders MC, van Leeuwen TG, Faber DJ. A literature review and novel theoretical approach on the optical properties of whole blood. Lasers Med Sci. 2014;29(2):453–79.

    Article  Google Scholar 

  47. Deisseroth K, Feng G, Majewska AK, Miesenböck G, Ting A, Schnitzer MJ. Next-generation optical technologies for illuminating genetically targeted brain circuits. J Neurosci. 2006;26(41):10380.

    Article  CAS  Google Scholar 

  48. Entcheva E. Cardiac optogenetics. American Journal of Physiology-Heart and Circulatory Physiology. 2013;304(9):H1179–91.

    Article  CAS  Google Scholar 

  49. Miesenböck G. The optogenetic catechism. Science. 2009;326(5951):395–9.

    Article  Google Scholar 

  50. Emiliani V, Entcheva E, Hedrich R, Hegemann P, Konrad KR, Lüscher C, et al. Optogenetics for light control of biological systems. Nature Reviews Methods Primers. 2022;2(1):55.

    Article  CAS  Google Scholar 

  51. Nagel G, Szellas T, Huhn W, Kateriya S, Adeishvili N, Berthold P, et al. Channelrhodopsin-2, a directly light-gated cation-selective membrane channel. Proc Natl Acad Sci. 2003;100(24):13940–5.

    Article  CAS  Google Scholar 

  52. Lin JY, Lin MZ, Steinbach P, Tsien RY. Characterization of engineered channelrhodopsin variants with improved properties and kinetics. Biophys J. 2009;96(5):1803–14.

    Article  CAS  Google Scholar 

  53. Levskaya A, Weiner OD, Lim WA, Voigt CA. Spatiotemporal control of cell signalling using a light-switchable protein interaction. Nature. 2009;461(7266):997–1001.

    Article  CAS  Google Scholar 

  54. Guntas G, Hallett RA, Zimmerman SP, Williams T, Yumerefendi H, Bear JE, et al. Engineering an improved light-induced dimer (iLID) for controlling the localization and activity of signaling proteins. Proc Natl Acad Sci. 2015;112(1):112–7.

    Article  CAS  Google Scholar 

  55. Kennedy MJ, Hughes RM, Peteya LA, Schwartz JW, Ehlers MD, Tucker CL. Rapid blue-light-mediated induction of protein interactions in living cells. Nat Methods. 2010;7(12):973–5.

    Article  CAS  Google Scholar 

  56. Zhou XX, Fan LZ, Li P, Shen K, Lin MZ. Optical control of cell signaling by single-chain photoswitchable kinases. Science. 2017;355(6327):836–42.

    Article  Google Scholar 

  57. Grusch M, Schelch K, Riedler R, Reichhart E, Differ C, Berger W, et al. Spatio-temporally precise activation of engineered receptor tyrosine kinases by light. Embo j. 2014;33(15):1713–26.

    Article  CAS  Google Scholar 

  58. Shimizu-Sato S, Huq E, Tepperman JM, Quail PH. A light-switchable gene promoter system. Nat Biotechnol. 2002;20(10):1041–4.

    Article  CAS  Google Scholar 

  59. Motta-Mena LB, Reade A, Mallory MJ, Glantz S, Weiner OD, Lynch KW, et al. An optogenetic gene expression system with rapid activation and deactivation kinetics. Nat Chem Biol. 2014;10(3):196–202.

    Article  CAS  Google Scholar 

  60. Chen X, Wang X, Du Z, Ma Z, Yang Y. Spatiotemporal control of gene expression in mammalian cells and in mice using the LightOn system. Curr Protoc Chem Biol. 2013;5(2):111–29.

    Article  CAS  Google Scholar 

  61. Renicke C, Schuster D, Usherenko S, Essen LO, Taxis C. A LOV2 domain-based optogenetic tool to control protein degradation and cellular function. Chem Biol. 2013;20(4):619–26.

    Article  CAS  Google Scholar 

  62. Niopek D, Benzinger D, Roensch J, Draebing T, Wehler P, Eils R, et al. Engineering light-inducible nuclear localization signals for precise spatiotemporal control of protein dynamics in living cells. Nat Commun. 2014;5(1):4404.

    Article  CAS  Google Scholar 

  63. Yumerefendi H, Lerner AM, Zimmerman SP, Hahn K, Bear JE, Strahl BD, et al. Light-induced nuclear export reveals rapid dynamics of epigenetic modifications. Nat Chem Biol. 2016;12(6):399–401.

    Article  CAS  Google Scholar 

  64. Yumerefendi H, Dickinson DJ, Wang H, Zimmerman SP, Bear JE, Goldstein B, et al. Control of protein activity and cell fate specification via light-mediated nuclear translocation. PLoS ONE. 2015;10(6): e0128443.

    Article  Google Scholar 

  65. Niopek D, Wehler P, Roensch J, Eils R, Di Ventura B. Optogenetic control of nuclear protein export. Nat Commun. 2016;7(1):10624.

    Article  CAS  Google Scholar 

  66. Dine E, Gil AA, Uribe G, Brangwynne CP, Toettcher JE. Protein phase separation provides long-term memory of transient spatial stimuli. Cell Syst. 2018;6(6):655-63.e5.

    Article  CAS  Google Scholar 

  67. Bracha D, Walls MT, Wei M-T, Zhu L, Kurian M, Avalos JL, et al. Mapping local and global liquid phase behavior in living cells using photo-oligomerizable seeds. Cell. 2018;175(6):1467-80.e13.

    Article  CAS  Google Scholar 

  68. Shin Y, Berry J, Pannucci N, Haataja MP, Toettcher JE, Brangwynne CP. Spatiotemporal control of intracellular phase transitions using light-activated optodroplets. Cell. 2017;168(1–2):159-71.e14.

    Article  CAS  Google Scholar 

  69. Guglielmi G, Barry JD, Huber W, De Renzis S. An optogenetic method to modulate cell contractility during tissue morphogenesis. Dev Cell. 2015;35(5):646–60.

    Article  CAS  Google Scholar 

  70. Kinjo T, Terai K, Horita S, Nomura N, Sumiyama K, Togashi K, et al. FRET-assisted photoactivation of flavoproteins for in vivo two-photon optogenetics. Nat Methods. 2019;16(10):1029–36.

    Article  CAS  Google Scholar 

  71. Schindler SE, McCall JG, Yan P, Hyrc KL, Li M, Tucker CL, et al. Photo-activatable Cre recombinase regulates gene expression in vivo. Sci Rep. 2015;5(1):13627.

    Article  Google Scholar 

  72. Qian Y, Cosio DMO, Piatkevich KD, Aufmkolk S, Su W-C, Celiker OT, et al. Improved genetically encoded near-infrared fluorescent calcium ion indicators for in vivo imaging. PLoS Biol. 2020;18(11): e3000965.

    Article  CAS  Google Scholar 

  73. Shemetov AA, Monakhov MV, Zhang Q, Canton-Josh JE, Kumar M, Chen M, et al. A near-infrared genetically encoded calcium indicator for in vivo imaging. Nat Biotechnol. 2021;39(3):368–77.

    Article  CAS  Google Scholar 

  74. Abdelfattah AS, Kawashima T, Singh A, Novak O, Liu H, Shuai Y, et al. Bright and photostable chemigenetic indicators for extended in vivo voltage imaging. Science. 2019;365(6454):699–704.

    Article  CAS  Google Scholar 

  75. Monakhov MV, Matlashov ME, Colavita M, Song C, Shcherbakova DM, Antic SD, et al. Screening and cellular characterization of genetically encoded voltage indicators based on near-infrared fluorescent proteins. ACS Chem Neurosci. 2020;11(21):3523–31.

    Article  CAS  Google Scholar 

  76. Watabe T, Terai K, Sumiyama K, Matsuda M. Booster, a red-shifted genetically encoded Förster Resonance Energy Transfer (FRET) biosensor compatible with cyan fluorescent protein/yellow fluorescent protein-based FRET biosensors and blue light-responsive optogenetic tools. ACS Sensors. 2020;5(3):719–30.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

YK is supported by a grant from Japan Society for the Promotion of Science; Grant-in-Aid for JSPS Fellows (20J01623), and Mochida Memorial Foundation for Medical and Pharmaceutical Research. KT is supported by a grant from Japan Society for the Promotion of Science (21H02715) and Research Grants of Princess Takamatsu Cancer Research Fund (PTCRF).

Author information

Authors and Affiliations

Authors

Contributions

YK participated in the drafting of the manuscript. KT reviewed the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Kenta Terai.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Konishi, Y., Terai, K. In vivo imaging of inflammatory response in cancer research. Inflamm Regener 43, 10 (2023). https://doi.org/10.1186/s41232-023-00261-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s41232-023-00261-x

Keywords