ArticlePDF Available

Tingible body macrophages

Authors:
418 © 2021 Journal of Oral and Maxillofacial Pathology | Published by Wolters Kluwer - Medknow
Described in 1885 by Flemming, tingible body
macrophages (TBMs) represent unique, large phagocytic
cells that reside in germinal centers (GCs) of secondary
lymphoid tissues and are a subset of mononuclear
phagocytes. TBMs contain many phagocytized, apoptotic
cells in various states of degradation. They owe their
name to their ability to actively phagocytose apoptotic
lymphoid cells, and the “tingible bodies” (TBs) observed
in their cytoplasm are apoptotic bodies.[1,2] They are seen
scattered among proliferating centroblasts, giving the
typical starry-sky appearance to developing GCs.[1,3]
The closely apposed medium sized tumor cells or the
normal lymphocytes in the reactive lymphnodes which
have minimal cytoplasm, and a dark nuclei impart a dark
blue background (the “sky”) to the histological sections.
These cells have a very high turnover rate, so that the
nearby macrophages get lled with cellular debris. Upon
xation, the cytoplasm of the macrophages disintegrates,
leaving round white spaces lled with debris (the “stars”),
imparting a “starry-sky pattern” at low magnication.
Nobody knows who created the histological comparison
with the “starry-sky” pattern. Perhaps, the unknown author
was inspired by the Dutch postimpressionist, Vincent
Willem van Gogh’s beautiful night paintings.[3]
“Starry-sky pattern” is distinctive for Burkitt lymphoma
but can be a nonspecific feature of pronounced
follicular hyperplasia seen in bone marrow, lymph node,
extranodal mass sections, some thymomas and rheumatoid
lymphadenopathy.[3] They may also be observed in other
malignant lymphomas with rapid cell turnover, such as
precursor B- or T-cell lymphomas and lymphoblastic
lymphomas. These high-grade lymphomas, however,
often accompany a monomorphic population of atypical
lymphoid cells as compared to the polymorphic pattern
of small “normal” lymphocytes in reactive conditions.[1]
Polarization, normal encompassing mantle zone, absence of
bcl-2 positivity, absence of capsular invasion and perinodal
fat invasion differentiate the reactive process in lymph
nodes from malignancy.[2,4] The appearance of TBMs and
a starry-sky appearance in the paracortex are indicative of
increased apoptosis and are suggestive of T-cell production
but may also occur with lymphocytolysis.[5] TBMs are also
noted in ne-needle aspiration of angiosarcoma of an
intraparotid lymph node.[6]
Lymphoid hyperplasia involving both the B-cell-rich
follicles and the T-cell-rich paracortex is generally a
reactive or immune response and is not considered to
be a preneoplastic lesion. Stimulated (reactive) follicles/
secondary follicles are usually larger than the unstimulated
primary follicles and will have a paler staining GC with
large lymphoblasts and increased number of apoptotic
lymphocytes and TBMs,[4] as seen in Figure 1.
TBMs range in size from 20 to 30 u or larger and contain
a variable number of inclusions [Figures 2 and 3]. Electron
microscopically TBMs show the nucleus, 12–15 p in
diameter, containing two dense, nucleolar-like areas and
peripheral dispersed chromatin. Mitochondria, smooth
and rough endoplasmic reticulum and a well-developed
Golgi region are present in the macrophage cytoplasm.[7]
The inclusions or the phagolysosomes (TBs) represent not
only nuclear but also cytoplasmic debris in varying stages
of lysis (apoptotic cell debris).[5,7] Following antigenic
stimulation, granulocytes are also seen in TBMs.[7]
It is generally accepted that the TBMs represent
phagocytized debris of small lymphocytes or both
lymphocyte and erythrocyte debris. This “graveyard
theory” emphasizes the phagocytosis of pyknotic small
lymphocytes. This theory was extended by Hamilton,
Trowell and Sundberg who postulated reutilization of
Figure 1: Photomicrograph from the reactive submandibular lymph
node of an elderly female patient with a large mandibular tumor,
histologically diagnosed as follicular ameloblastoma. (a) Capsule,
(b) Paracortical germinal center. Tingible body macrophages impart
the paracortical germinal center “starry-sky” appearance (H&E, ×10)
Tingible body macrophages
Enigmatic Morphoinsight
Gotur and Wadhwan: Tingible body macrophages
Journal of Oral and Maxillofacial Pathology | Volume 24 | Issue 3 | September-December 2020 419
small lymphocytes in lymphocytopoiesis. Andrew stressed
the role of degeneration and phagocytosis of small
lymphocytes in GCs as indicating that these centers are
not germinal but reactive, contradicting Hellman. Ortega
and Mellors suggested that intrinsic GC cells degenerated
after secreting gamma globulin and were phagocytized by
TBMs. Electron microscopically plasma cell inclusion by
TBs is also seen in hyperplastic GCs of lymphatic tissue in
the mouse following antigenic stimulation. The plasma cell
phagocytosis by TBMs may reect plasma cell proliferation
rather than lymphocyte production from the population
of cells comprising GCs after antigenic stimulation. This
proposal is in agreement with the suggestion by Ringertz
and Adamson and Congdon and Goodman that the
centers form either antibody-producing plasma cells or
lymphocytes, depending on an antigenic stimulus. This
idea is also consistent with the functional proposal by
Ortega and Mellors that depleted GC cells are phagocytized
following protein synthetic activity.[7]
Unique to TBMs is the GC microenvironment,
characterized functionally by long-term antigen retention
on follicular dendritic cells, antigen presentation by
B-cells to T-helper cells, a high rate of somatic mutations,
affinity maturation, induction of antibody-forming
cells and memory B-cell development. Apart from the
scavenging function, TBMs also may be important in
initiating the GC reaction. This suggestion was based on
the observations that TBM initially appeared at the onset
of GC development and that a peak number of TBMs
were seen at peak GC development. However, it has been
shown that GCs can develop in old mice in the absence of
TBM. This advocates a regulatory role of TBM and that
TBMs are more likely to downregulate than to stimulate
the GC reaction.[8] Further, TBMs were found to be rich
in prostaglandins via which they downregulate the GC
reaction.[2,9]
It has been suggested that the scavenging activity of TBM
may play a signicant role in preventing autoimmunity.[2]
Fat globule-epidermal growth factor 8 (Mfge8) promotes
TBMs to engulf apoptotic bodies in GCs and helps
minimize autoimmunity.[9] Furthermore, this is illustrated
by a confocal microscopic study of TBMs, to evaluate the
in vivo capacity to remove apoptotic cell material, conducted
on patients with systemic lupus erythematosus (SLE). In
lymph nodes GCs from the patients with SLE, the apoptotic
cells were more and the TBMs were reduced in number, in
contrast to the control group. This nding suggested that
in SLE patients, apoptotic cells are not properly cleared
by TBMs. Consequently, retained nuclear autoantigens can
be presented to and bind to follicular dendritic cells and
may thus provide survival signals for autoreactive B-cells,
thereby initiating or propagating autoimmune disease.[2,10]
To conclude, TBMs in GCs of reactive lymph nodes
represent a benign process which should be differentiated
from the lymphomas and other malignancies. Further
studies are required to understand the role of TBMs in the
pathogenesis of various autoimmune diseases.
Financial support and sponsorship
Nil.
Conflicts of interest
There are no conicts of interest.
Suhasini Palakshappa Gotur, Vijay Wadhwan
Department of Oral Pathology and Microbiology, Subhar Dental College
Figure 2: Higher magnication of Figure 1, showing (a) tingible body
macrophages with intracytoplasmic apoptotic bodies, which represent
nuclear debris, surrounded by (b) lymphocytes (H&E, ×40)
Figure 3: Hand-drawn illustration showing (a) tingible body
macrophages with intracytoplasmic apoptotic bodies, surrounded by
(b) lymphocytes
Gotur and Wadhwan: Tingible body macrophages
420 Journal of Oral and Maxillofacial Pathology | Volume 24 | Issue 3 | September-December 2020
and Hospital, Swami Vivekanand Subhar University, Meerut, Uar
Pradesh, India
E-mail: suhasrajukk@gmail.com
Submied: 27-Jul-2020,
Accepted: 06-Oct-2020, Published: 09-Jan-2021
REFERENCES
1. Tingible Body Macrophage - an overview _ ScienceDirect Topics.
Available from: https://www.sciencedirect.com/topics/ biochemistry
genetics and molecular biology/tingible body macrophage. [Last
accessed on 2020 May 30].
2. North JP, McCalmont TH. Angiosarcoma with tingible body
macrophages. J Cutan Pathol 2011;38:683.
3. Andrade-Filho Jde S. Analogies in medicine: Starry-sky appearance. Rev
Inst Med Trop Sao Paulo 2014;56:541-2.
4. Elmore SA. Histopathology of the lymph nodes. Toxicol Pathol
2006;34:425-54.
5. Willard-Mack CL. Normal structure, function, and histology of lymph
nodes. Toxicol Pathol 2006;34:409-24.
6. Ducharne-Asuaje E, Dorion D, Lamarre L, Coindre JM, Geha S. Primary
high-grade poorly differentiated angiosarcoma of an intra-parotid lymph
node. Head Neck Pathol 2016;10:225-8.
7. Swartzendruber DC, Congdon CC. Electron microscope observations
on tingible body macrophages in mouse spleen. J Cell Biol 1963;19:641-6.
8. Smith JP, Burton GF, Tew JG, Szakal AK. Tingible body macrophages
in regulation of germinal center reactions. Dev Immunol 1998;6:285-94.
9. El Shikh ME, Pitzalis C. Follicular dendritic cells in health and disease.
Front Immunol 2012;3:292.
10. Baumann I, Kolowos W, Voll RE, Manger B, Gaipl U, Neuhuber WL,
et al. Impaired uptake of apoptotic cells into tingible body macrophages
in germinal centers of patients with systemic lupus erythematosus.
Arthritis Rheum 2002;46:191-201.
Access this article online
Quick Response Code:
Website:
www.jomfp.in
DOI:
10.4103/jomfp.JOMFP_314_20
This is an open access journal, and arcles are distributed under the terms of the Creave
Commons Aribuon-NonCommercial-ShareAlike 4.0 License, which allows others to
remix, tweak, and build upon the work non-commercially, as long as appropriate credit
is given and the new creaons are licensed under the idencal terms.
How to cite this article: Gotur SP, Wadhwan V. Tingible body macrophages.
J Oral Maxillofac Pathol 2020;24:418‑20.
... ; https://doi.org/10.1101/2022.08.18.504434 doi: bioRxiv preprint TBMs contain apoptotic cellular debris at different degradation stages and are named after apoptotic nuclear debris ('tingible bodies') that can be observed in their cytoplasm. A presence of TBMs rich in tingible bodies is a hallmark of follicular hyperplasia and Burkitt's lymphoma, both characterized by fast cell turnover (Gotur and Wadhwan 2020). We examined hyperplastic LN from two different patients and two Burkitt's lymphoma cases, and found that TBMs in both conditions contain many tingible bodies and display high IL4I1 expression (Fig 4C-D). ...
Preprint
Full-text available
Macrophages are the most abundant immune cell type in the tumor microenvironment (TME). Yet the spatial distribution and cell interactions that shape macrophage function are incompletely understood. Here we use single-cell RNA sequencing data and multiplex imaging to discriminate and spatially resolve macrophage niches within benign and malignant breast and colon tissue. We discover four distinct tissue-resident macrophage (TRM) layers within benign bowel, two TRM niches within benign breast, and three tumor-associated macrophage (TAM) populations within breast and colon cancer. We demonstrate that IL4I1 marks phagocytosing macrophages, SPP1 TAMs are enriched in hypoxic and necrotic tumor regions, and a novel subset of FOLR2 TRMs localizes within the plasma cell niche. Furthermore, NLRP3 TAMs that colocalize with neutrophils activate an inflammasome in the TME and in Crohn's disease and are associated with poor outcomes in breast cancer patients. This work suggests novel macrophage therapy targets and provides a framework to study human macrophage function in clinical samples.
Article
Full-text available
Avian influenza viruses (AIV) of the H7N7 subtype are enzootic in the wild bird reservoir in Europe, cause infections in poultry, and have sporadically infected humans. The non-structural protein PB1-F2 is encoded in a second open frame in the polymerase segment PB1 and its sequence varies with the host of origin. While mammalian isolates predominantly carry truncated forms, avian isolates typically express full-length PB1-F2. PB1-F2 is a virulence factor of influenza viruses in mammals. It modulates the host immune response, causing immunopathology and increases pro-inflammatory responses. The role of full-length PB1-F2 in IAV pathogenesis as well as its impact on virus adaptation and virulence in poultry remains enigmatic. Here, we characterised recombinant high pathogenicity AIV (HPAIV) H7N7 expressing or lacking PB1-F2 in vitro and in vivo in chickens. In vitro, full-length PB1-F2 modulated viability of infected chicken fibroblasts by limiting apoptosis. In chickens, PB1-F2 promoted gastrointestinal tropism, as demonstrated by enhanced viral replication in the gut and increased cloacal shedding. PB1-F2’s effects on cellular immunity however were marginal. Overall, chickens infected with full-length PB1-F2 virus survived for shorter periods, indicating that PB1-F2 is also a virulence factor in bird-adapted viruses. Supplementary Information The online version contains supplementary material available at 10.1186/s13567-023-01257-8.
Article
As we explore other planetary bodies, astronauts will face unique environmental and physiological challenges. The human immune system has evolved under Earth’s gravitational force. Consequently, in the microgravity environment of space, immune function is altered. This can pose problematic consequences for astronauts on deep space missions where medical intervention will be limited. Studying the unique environment of microgravity has its challenges, yet current research has uncovered immunological states that are probable during exploration missions. As microgravity-induced immune states are uncovered, novel countermeasure developments and personalized mitigation programs can be designed to improve astronaut health. This can also benefit immune-related monitoring programs for disorders on Earth. This is a comprehensive review, including gaps in knowledge, of simulated and spaceflight microgravity studies in human and rodent models.
Preprint
Full-text available
Tumor-associated macrophages (TAMs) display heterogeneous phenotypes. Yet the exact tissue cues that shape macrophage functional diversity are incompletely understood. Here we discriminate, spatially resolve and reveal the function of five distinct macrophage niches within malignant and benign breast and colon tissue. We found that SPP1 TAMs reside in hypoxic and necrotic tumor regions, and a novel subset of FOLR2 tissue-resident macrophages (TRMs) supports the plasma cell tissue niche. We discover that IL4I1 macrophages populate niches with high cell turnover where they phagocytose dying cells. Significantly, IL4I1 TAMs abundance correlates with anti-PD1 treatment response in breast cancer. Furthermore, NLRP3 inflammasome activation in NLRP3 TAMs correlates with neutrophil infiltration in the tumors and is associated with poor outcome in breast cancer patients. This suggests the NLRP3 inflammasome as a novel cancer immunotherapy target. Our work uncovers context-dependent roles of macrophage subsets and suggests novel predictive markers and macrophage subset-specific therapy targets.
Article
Full-text available
Mature B cell malignancies constitute a wide range of biologically and clinically heterogeneous hematological diseases. Despite an increasingly thorough understanding of the pathophysiology of these pathologies and significant improvements in therapies, a dismal outcome still affects a large number of patients. Therefore, further investigations into new treatment perspectives are highly needed and they depend entirely on the ex vivo culture of patient cells. Primary cells usually demand superior culture models, as they are notoriously difficult to cultivate. The literature is not devoid of approaches ranging from two- to three-dimensional systems for culturing mature malignant primary B cells. However, they display substantial protocol inter-variation. This imposes a high risk of failures, repeats, and inconsistent results, which are neither compatible with the rare value of primary cells nor the efficiency of the drug discovery process. In this review, we provide a thorough overview of the different approaches that have been implemented in the literature for the culture of mature malignant primary B cells, and we discuss associated considerations and limitations to assist researchers in determining a fit-for-purpose culture system, thereby attempting to reduce the number of trials and errors as well as associated biomaterial expenditure.
Article
Full-text available
Follicular dendritic cells (FDCs) are unique immune cells that contribute to the regulation of humoral immune responses. These cells are located in the B-cell follicles of secondary lymphoid tissues where they trap and retain antigens (Ags) in the form of highly immunogenic immune complexes (ICs) consisting of Ag plus specific antibody (Ab) and/or complement proteins. FDCs multimerize Ags and present them polyvalently to B-cells in periodically arranged arrays that extensively crosslink the B-cell receptors for Ag (BCRs). FDC-FcγRIIB mediates IC periodicity, and FDC-Ag presentation combined with other soluble and membrane bound signals contributed by FDCs, like FDC-BAFF, -IL-6, and -C4bBP, are essential for the induction of the germinal center (GC) reaction, the maintenance of serological memory, and the remarkable ability of FDC-Ags to induce specific Ab responses in the absence of cognate T-cell help. On the other hand, FDCs play a negative role in several disease conditions including chronic inflammatory diseases, autoimmune diseases, HIV/AIDS, prion diseases, and follicular lymphomas. Compared to other accessory immune cells, FDCs have received little attention, and their functions have not been fully elucidated. This review gives an overview of FDC structure, and recapitulates our current knowledge on the immunoregulatory functions of FDCs in health and disease. A better understanding of FDCs should permit better regulation of Ab responses to suit the therapeutic manipulation of regulated and dysregulated immune responses.
Article
Full-text available
Tingible body macrophages (TBM), long thought simply as scavengers of apoptotic lymphocytes, are located in the unique microenvironment of germinal centers in close proximity to antigen-retaining follicular dendritic cells (FDC). Observations that TBM endocytose FDC-iccosomal (immune-complex coated bodies) antigen suggested that TBM might present this antigen and help regulate the germinal center reaction. To test for antigen presentation, the ovalbumin (OVA)-specific T(H) hybridoma, 3DO-54.8, which produces IL-2 on receiving effective presentation of OVA, were used as responders to OVA-bearing TBM. Results showed that OVA-bearing TBM failed to induce IL-2 production. Furthermore, addition of TBM to IL-2-inducing positive controls (B cells) not only failed to augment IL-2 production, but rather TBM significantly (55-90%) reduced B-cell induction of IL-2. We found that TBM were rich in prostaglandin by comparison with other nongerminal center lymph node macrophages and that addition of indomethacin to the cultures reversed the inhibitory effect of TBM. Depletion of TBM from enriched preparations, prior to addition to positive control cultures, also abrogated the inhibitory effect on IL-2 production. These data support the concept that TBM, within the unique microenvironment of germinal centers, may be specialized to downregulate the germinal center reaction.
Article
Full-text available
Lymph nodes function as filters of tissues and tissue fluids and are sites of origin and production of lymphocytes for normal physiological functions. As part of this normal function, they react to both endogenous and exogenous substances with a variety of specific morphological and functional responses. Lesions can be both proliferative and nonproliferative, and can be treatment-related or not. The histological evaluation of lymph nodes is necessary in order to understand the immunotoxic effects of chemicals with the resulting data providing an important component of human risk assessment. It is the challenge of the toxicologic pathologist to interpret the pathology data within the complete clinical evaluation of the entire animal. Daily insults, ageing and toxins can alter the normal histology and primary function of lymph nodes. Therefore it is important to distinguish and differentiate lesions that occur naturally during normal development and ageing from those that are induced by xenobiotics. To achieve this goal, comparison with strain- age- and sex-matched controls is crucial.
Article
To investigate the fate of apoptotic cells in the germinal centers (GCs) of patients with systemic lupus erythematosus (SLE).
Article
Head and neck angiosarcoma is an infrequent malignant vascular tumor most commonly found in the skin and soft tissue of the head and neck. Most head and neck angiosarcomas are metastatic to cervical lymph nodes from other primitive location. We describe herein a case of primary high-grade poorly differentiated angiosarcoma arising in an intra-parotid lymph node, discuss the value of immunohistochemical stains for differential diagnosis, and review the literature concerning head and neck angiosarcoma. A 47-year-old man presented with a painless mass that had grown for a period of 6 months in the parotid area. The CT-scan revealed a left parotid lesion of 17 mm. Fine needle aspiration was considered suspicious for lymphoma or poorly differentiated carcinoma. A superficial parotidectomy was performed. On gross examination, the lesion was a well-defined, gray, homogeneous mass of 15 mm of diameter. Microscopic examination showed a normal parotid tissue and a poorly differentiated malignant neoplasm in an intra-parotid lymph node. The tumor had a pseudo-alveolar pattern, with large pleomorphic epithelioid cells, abundant eosinophilic cytoplasm, large vesicular nuclei, and one or more prominent nucleoli. Atypical mitoses were seen. Neoplastic malignant cells stained positive for Vimentin, CD31, D2-40, factor VIII, ERG, and partially for CD34. A positron emission tomography scan was made to search for a primary neoplasia, but no other tumor was localized. The diagnosis of primary high-grade, poorly differentiated, intra-parotid lymph node angiosarcoma was established.
Article
To investigate the fate of apoptotic cells in the germinal centers (GCs) of patients with systemic lupus erythematosus (SLE). Lymph node biopsy specimens obtained from 7 SLE patients with benign follicular hyperplasia, 5 non-SLE patients with benign follicular hyperplasia (non-SLE), 5 patients with malignant follicular lymphoma, and 3 patients with dermatopathic lymphadenitis were stained with monoclonal antibodies against macrophages (CD68) and follicular dendritic cells (CR2/CD21). TUNEL staining and transmission electron microscopy were performed to detect apoptotic cells. Confocal microscopy was used to evaluate the in vivo capacity of tingible body macrophages to remove apoptotic cell material. In a subgroup of patients with SLE, apoptotic cells accumulated in the GCs of the lymph nodes. The number of tingible body macrophages, which usually contained engulfed apoptotic nuclei, was significantly reduced in these patients. In contrast to what was observed in all controls, TUNEL-positive apoptotic material from SLE patients was observed to be directly associated with the surfaces of follicular dendritic cells (FDCs). Our findings suggest that in a sub-group of SLE patients, apoptotic cells are not properly cleared by tingible body macrophages of the GCs. Consequently, nuclear autoantigens bind to FDCs and may thus provide survival signals for autoreactive B cells. This action may override an important control mechanism for B cell development, resulting in the loss of tolerance for nuclear antigens.
Article
Lymph nodes are traditionally regarded as having three compartments, the cortex, paracortex and medulla. B and T cells home to separate areas within these compartments, interact with antigen presenting cells, and undergo clonal expansion. This paper provides structural and functional details about how the lymph node brings lymphocytes and antigen presenting cells together. The concept of the lymphoid lobule as the basic functional and anatomic unit of the lymph node is developed and utilized to provide a framework for understanding lymph node pathobiology. Understanding the histomorphologic features of the lymphoid lobule and the role of the reticular meshwork scaffolding of the lymph node and how these related to the cortex, paracortex and medulla provides a unique approach to understanding lymph node structure and function.