A consensus system for classification of mouse lymphoid neoplasms according to their histopathologic and genetic features has been an elusive target for investigators involved in understanding the pathogenesis of spontaneous cancers or modeling human hematopoietic diseases in mice. An international panel of scientists with expertise in mouse and human hematopathology joined with the hematopathology subcommittee of the Mouse Models for Human Cancers Consortium to develop criteria for definition and classification of these diseases together with a standardized nomenclature. The fundamental elements contributing to the scheme are clinical features, morphology, immunophenotype, and genetic characteristics. The resulting classification has numerous parallels to the World Health Organization classification of human lymphoid tumors while recognizing differences that may be species specific. The classification should facilitate communications about mouse models of human lymphoid diseases.

A systematic classification of mouse hematopoietic neoplasms was first put forward by Dunn in 1954.1 In this assemblage, tumor types were related to presumed cells of origin, including undifferentiated cells, lymphocytes, granulocytes, “reticulum cells” Types A and B, plasmacytes, and tissue mast cells. Similar names for tumor types can be found in the 1966 Rappaport nomenclature for human hematopoietic tumors.2 Following the identification of T- and B-cell lymphocyte subsets in the 1970s, Pattengale and Taylor3revised the mouse classification scheme to generally parallel the 1974 Lukes-Collins proposal for human hematopoietic neoplasms.4 Within these frameworks, most tumors previously defined as of reticulum cell origin were recognized to correspond to B-cell lineage lymphomas. In 1994, Fredrickson et al5 used the human Kiel classification of 19816as modified in 19887 as the basis for a nomenclature. Among other features, it encompassed previously unrecognized subtypes of mouse B-cell lineage lymphoma, including splenic marginal zone lymphoma (SMZL).8 Most recently, the consensus nomenclatures for human hematopoietic tumors established by the 1995 Revised European-American Lymphoma9 and descendant 2001 World Health Organization (WHO) schemes10,11 were adopted as a model by investigators from the National Institutes of Health in an effort to develop a contemporaneous scheme for classification of mouse lymphoma and leukemia.12-14 

The iterative nature of the lists and definitions of disease entities in the mouse derives from the increasingly well-supported presumption that carefully defined and validated model systems can provide fundamental insights into human disorders with attendant implications for prevention and intervention. The opportunities to extend this concept provided by manipulation of the mouse genome were recognized in the formation of the Mouse Models of Human Cancers Consortium (MMHCC) by the United States National Cancer Institute (http://emice.nci.nih.gov). The Hematopathology Subcommittee of the MMHCC was given the charge of developing a consensus list of hematopoietic neoplasms with descriptions and criteria for diagnosis. The goal was to define disease entities that could be recognized by pathologists and related to human disorders where possible. To meet this challenge, gatherings of international experts in human and mouse hematopathology were convened. The result of their deliberations is a proposed classification of hematopoietic diseases that stratifies disorders according to cell lineage. The resulting major subgroups are, accordingly, lymphoid and nonlymphoid. The lymphoid disorders will be discussed here and the nonlymphoid in a separate article.48 

Most of our present knowledge of mouse hematopoietic neoplasms is based on studies of such diseases in inbred and outbred mice occurring either spontaneously or following induction with irradiation, chemicals, or exogenous infection with murine leukemia viruses (MuLVs).3,5,8,15,16 There is considerable strain dependence of disease types and incidence, with much of the differences being genetically determined. Some of these variations are associated with expression of endogenous MuLV and virus-controlling genes,17,18 although infectious MuLVs are not required for induction or progression of many disorders. These considerations are not obviated for diseases developing in genetically engineered mice (GEM)19 but are likely of less importance. Although the range of lymphoid neoplasms seen in conventional mice is broad, it has been substantially extended through genetic engineering. Some of the newly recognized disorders in GEM recapitulate those of humans with greater or lesser levels of fidelity, whereas others appear not to. Indeed, some of the lymphomas and leukemias seen in GEM have never been seen previously as spontaneous lymphomas in mice. In this regard, it is important to remember that only a limited number of strains have been evaluated in a rigorous manner for the characteristics of spontaneous lymphomas. The information at hand that forms much of the basis for our proposals may thus not be representative of the full range of diseases that develop among unmanipulated mice.

A prominent goal of studying mouse tumors is to develop an understanding of pathogenesis that would provide opportunities for preventing or treating similar disorders of humans. It has, therefore, been important to determine whether diseases in the 2 species are true homologs and deserve identical names. We have found this case is often difficult to make. The reasons are multiple. Prominent among these reasons are fundamental differences between the species in the characteristics of primary and secondary lymphoid organs. Uniquely in mice, extramedullary hematopoiesis continues in the red pulp of the spleen throughout life, and the thymus persists well into adulthood. The splenic marginal zone of mice has also been shown to differ significantly from that in humans.20 

An additional critical difference is that many modalities used to classify human lymphomas have been applied much less routinely or rigorously to studies of mouse tumors. Without these direct comparisons at hand, guesswork and wishful thinking provide insufficient grounds for identifying true parallels. Consequently, although the committee developed a set of recommendations to be used by pathologists and investigators diagnosing these diseases, these proposals will be altered and updated as additional information becomes available. The fundamental elements contributing to classification of lymphomas in the WHO classification are clinical features, morphology, immunophenotype, and genetic abnormalities. The committee concluded that the same types of information should be developed, when possible, in studies of mouse hematopoietic neoplasms.

Committee recommendations for approaches to diagnosing lymphoid neoplasms of mice are not presented here because of space constraints but are provided in supplementary material available on theBlood website; see the Supplemental Data link at the top of the online article. These recommendations should be reviewed in detail by those new to the field of mouse hematopathology studying either spontaneous or induced models of disease. The guidelines may also provide a helpful review for more established investigators. We anticipate that more uniform application of these approaches to diagnosis and classification by the hematopathology community will facilitate the characterization of lymphomas and leukemias and yield descriptions that can be more readily understood by all. The supplementary material provides approaches to necropsies, sample collection and storage, fixation and staining, as well as flow cytometric and molecular evaluations of tumor samples. Critical parameters for distinguishing reactive from malignant processes complete the online package. The terminology used in the textual material of the print version is based on the definitions provided in the supplementary information.

The consensus recommendations of the Hematopathology Subcommittee of the MMHCC for classification of mouse lymphoid neoplasms follow the WHO classification in many respects but use distinct terminology when appropriate. Differences in the schemes have several origins that will be dealt with in describing each of the diseases.

Just as the classification of human lymphomas is a work in progress with the WHO scheme being just the most recent version, we view the proposed classification for mouse lymphoid neoplasms as a statement of where we are at present. The scheme presented in Figure1 will be revised as new data on established diseases, both human and mouse, become available and as new disorders are described.

Fig. 1.

Proposed classification of mouse lymphoid neoplasms.

Fig. 1.

Proposed classification of mouse lymphoid neoplasms.

Close modal

Table 1 compares the features of the lymphoma/leukemia types and their relation to diseases in humans. Several conventions will be followed in the table. First, the phenotype of mature B-cell lymphomas will be given as sIg+ B220 [CD45R(B220)]+ CD19+ with the recognition that other markers may be useful in distinguishing distinct types. Second, the molecular characteristics of mature B-cell lymphomas, regardless of type, will be presented in Table 1 as if both heavy chain and both kappa light chain alleles have undergone rearrangements [IgH R/R, IgK R/R] with the recognition that only one allele of each may be rearranged and that lambda light chain may be rearranged and used in a subset of the neoplasms. In these diagnostic categories, the T-cell locus will be listed as unrearranged, indicating only the T-cell receptor locus (TCRβ G/G), with the recognition that the locus is rearranged in some B-lineage tumors. An expanded description of the occurrence of lymphomas of each diagnostic category is given in Table 5 of the supplementary material, and a more complete listing will be given at the MMHCC Web site (http://mmr.afs.apelon.com/heme/). Finally, although cytologic details are described, they pertain to the appearance of cells in hematoxylin and eosin (H&E)–stained tissue sections of formalin-fixed, paraffin-embedded tissues. Representative cases of lymphoma types are shown in Figures 2-4.

Fig. 2.

B-cell lymphomas.

(A) Small B-cell lymphoma. A uniform population of small, mature, round lymphocytes within an expanded white pulp with similar cells invading the red pulp (H&E, × 750). (B) Splenic marginal zone lymphoma. Widening of the marginal zone with medium-sized B cells containing prominent eosinophilic cytoplasm with no red pulp infiltration (H&E, × 400). (C) Advanced SMZL. Evidenced by invasion of the red pulp and compression of the white pulp (H&E, × 400). (D) Advanced SMZL. Centroblastlike cells with mitotic figures obliterating normal architecture. Cytoplasm is moderately extensive (H&E, × 750). (E) Follicular B-cell lymphoma. The pale zone represents loss of small dark lymphocytes in both the periarteriolar lymphoid sheath (PALS) and follicle and replacement with mixed centrocytes and centroblasts (H&E, × 150). (F) Follicular B-cell lymphoma. Mixed population of large centroblasts, cleaved centrocytes, and small lymphocytes with few immunoblasts (H&E, × 750).

Fig. 2.

B-cell lymphomas.

(A) Small B-cell lymphoma. A uniform population of small, mature, round lymphocytes within an expanded white pulp with similar cells invading the red pulp (H&E, × 750). (B) Splenic marginal zone lymphoma. Widening of the marginal zone with medium-sized B cells containing prominent eosinophilic cytoplasm with no red pulp infiltration (H&E, × 400). (C) Advanced SMZL. Evidenced by invasion of the red pulp and compression of the white pulp (H&E, × 400). (D) Advanced SMZL. Centroblastlike cells with mitotic figures obliterating normal architecture. Cytoplasm is moderately extensive (H&E, × 750). (E) Follicular B-cell lymphoma. The pale zone represents loss of small dark lymphocytes in both the periarteriolar lymphoid sheath (PALS) and follicle and replacement with mixed centrocytes and centroblasts (H&E, × 150). (F) Follicular B-cell lymphoma. Mixed population of large centroblasts, cleaved centrocytes, and small lymphocytes with few immunoblasts (H&E, × 750).

Close modal
Fig. 3.

B-cell lymphomas.

(A) DLBCL-centroblastic. A uniform population of large centroblasts with nuclear membrane-associated nucleoli has completely replaced normal splenic structure (H&E, × 750). (B) DLBCL-immunoblastic. A pleomorphic population of immunoblasts with large nucleoli as well as some centroblasts (H&E, × 750). (C) DLBCL-histiocyte associated. A pleomorphic population of large lymphoid cells with prominent area of histiocytic cells (lower left) (H&E, × 1000). (D) DLBCL-primary mediastinal (thymic). Tumor arising in the thymus composed of a mixed population of B220+ cells (inset; H&E, × 15) including immunoblasts (H&E, × 750). (E) Classic Burkitt lymphoma. Large cells with prominent nucleoli central or in peripheral areas of the nucleus. Apoptosis is a striking feature (H&E, × 1000). (F) Burkittlike lymphoma. Medium-sized cells with central nucleoli or stipple chromatin with moderate apoptosis (H&E, × 750).

Fig. 3.

B-cell lymphomas.

(A) DLBCL-centroblastic. A uniform population of large centroblasts with nuclear membrane-associated nucleoli has completely replaced normal splenic structure (H&E, × 750). (B) DLBCL-immunoblastic. A pleomorphic population of immunoblasts with large nucleoli as well as some centroblasts (H&E, × 750). (C) DLBCL-histiocyte associated. A pleomorphic population of large lymphoid cells with prominent area of histiocytic cells (lower left) (H&E, × 1000). (D) DLBCL-primary mediastinal (thymic). Tumor arising in the thymus composed of a mixed population of B220+ cells (inset; H&E, × 15) including immunoblasts (H&E, × 750). (E) Classic Burkitt lymphoma. Large cells with prominent nucleoli central or in peripheral areas of the nucleus. Apoptosis is a striking feature (H&E, × 1000). (F) Burkittlike lymphoma. Medium-sized cells with central nucleoli or stipple chromatin with moderate apoptosis (H&E, × 750).

Close modal
Fig. 4.

Plasma cell and T-cell lymphomas.

(A) Plasmacytoma. Well-differentiated neoplasm with progression from large plasmablasts with prominent central nucleoli to mature plasma cells (H&E, × 750). (B) Plasmacytoma. Moderately differentiated with maturation of plasmablasts into plasma cells (H&E, × 750). (C) Anaplastic plasmacytoma. Large blast cells with plentiful amphophilic cytoplasm (H&E, × 750). (D) Precursor T-cell lymphoblastic lymphoma. Uniform population of medium-sized cells with central or small scattered nucleoli (H&E, × 1000). (E) T-natural killer cell leukemia. Mature small lymphocytes and some large immature lymphoblasts (H&E, × 750). (F) Large cell anaplastic T-cell lymphoma. Comprising mainly large anaplastic immunoblastlike cells with large nucleoli (H&E, × 750).

Fig. 4.

Plasma cell and T-cell lymphomas.

(A) Plasmacytoma. Well-differentiated neoplasm with progression from large plasmablasts with prominent central nucleoli to mature plasma cells (H&E, × 750). (B) Plasmacytoma. Moderately differentiated with maturation of plasmablasts into plasma cells (H&E, × 750). (C) Anaplastic plasmacytoma. Large blast cells with plentiful amphophilic cytoplasm (H&E, × 750). (D) Precursor T-cell lymphoblastic lymphoma. Uniform population of medium-sized cells with central or small scattered nucleoli (H&E, × 1000). (E) T-natural killer cell leukemia. Mature small lymphocytes and some large immature lymphoblasts (H&E, × 750). (F) Large cell anaplastic T-cell lymphoma. Comprising mainly large anaplastic immunoblastlike cells with large nucleoli (H&E, × 750).

Close modal

Hypermutation of immunoglobulin variable region sequences indicates that human lymphomas of this type are of germinal center B-cell origin or have evidence of having passaged the germinal center. Careful studies among medical hematopathologists led to the conclusion that it is difficult to reproducibly distinguish morphologic variants designated centroblastic, immunoblastic, T-cell/histiocyte rich, and other subsets of this disorder defined in pre-WHO nomenclatures.9 The prospect that morphologic variants may in time be associated with specific genetic features defined by using complementary DNA microarray or other technologies led to the suggestion that use of the terminology for the variants remains optional.10 The mouse lymphomas may provide a stronger case for continuing with subset designations in view of highly distinctive morphologic and histologic features of several proposed types (Figure 3A-D). Until such time as these designations can be buttressed by criteria other than histologic appearance, it would seem prudent to keep the use of these morphologic subgroups optional.

There are aspects to distinguishing diffuse large B-cell lymphomas (DLBCLs) from other entities in both humans and mice that deserve comment. First, human follicular lymphomas are defined by a combination of what, for purposes of discussion, can be termed cytologic and architectural features within lymph nodes. The cytologic range of these neoplasms comprises primarily centrocytes and centroblasts. The representation of centroblasts is variable, ranging from less than 5 per high-power field to solid sheets of blasts. The architectural hallmark of this disease is poorly defined follicular structures. The proportion of lymph nodes with follicular structures can vary from less than 25% to complete, the remaining areas being diffusely involved. Retention of follicular architecture may thus be said to be the signature distinction between human DLBCL of centroblastic type and follicular lymphomas comprising exclusively centroblasts.

In mice, the cytologic and architectural features that distinguish follicular from DLBCLs are distinct. First, mouse follicular tumors composed almost uniformly of centrocytes are extremely rare, and more evenly balanced populations of centrocytes and centroblasts tend to be the distinguishing feature (Figure 2E). Second, follicular lymphomas in mice usually arise in spleen rather than lymph nodes, meaning that the architectural-distinguishing features of the diseases are fundamentally different for the 2 species. The appearance of involved spleen is nodular at both the gross and microscopic levels with progressive expansion of multiple or all follicles in the white pulp leading to compression of the red pulp as well as the T-cell periarteriolar lymphocytic sheath. Progression may lead to lymphomas with an almost pure population of centroblasts that remain restricted to the white pulp or spread into the red pulp but with the follicular origin still evident. These are centroblastic lymphomas of follicular origin. For historical reasons, it is worth noting that Pattengale and Taylor3 defined follicular center cell lymphomas as small cell, large cell, and mixed. The large cell type corresponds to what we are here calling DLBCL.

Two other subtypes of centroblastic lymphoma have been observed. The first represents a progression of SMZL to a higher grade with clear anatomic evidence of its origins.8 The other type comprises centroblastic tumors for which an origin from the follicle or marginal zone cannot be inferred.

Recent studies have described clonal B-cell lymphomas with high proportions of nonclonal T cells.21 It has been suggested that these mouse tumors may be the equivalent of human T-cell–rich DLBCL, but the frequencies of T cells have not been determined by flow cytometry and may not reach the levels seen in the human disease.

Second, there are mouse lymphomas that are histologically indistinguishable from precursor B-cell and precursor T-cell lymphomas but comprise mature sIg+ B cells.22,23 In the veterinary literature, these lymphomas have been classified together with precursor neoplasms as lymphoblastic lymphoma. The fact that some lymphomas of this type derived from 2 different populations of mice exhibited structural alterations in BCL-6, the most common genetic abnormality of human DLBCL,24 originally was used to argue for their classification as DLBCL, despite their lymphoblastic appearance and the lack of human tumors with this combination of histologic and immunophenotypic features.22,23 With the more recent opportunity to study large numbers of cases of mouse Burkitt lymphoma (Figure 3E),25 it became apparent that a proportion of the sIg+ lymphoblastic tumors resembled mouse Burkitt lymphoma histologically while sharing a proliferative fraction approaching 100%. This finding suggests atypical Burkitt or Burkittlike as perhaps more appropriate terminology for these lymphomas (Figure 3F). In the WHO classification, however, this term is reserved for cases with nonclassical cytology despite proven or likely translocations activating MYC.10 MYC is not rearranged in a significant proportion of the mouse lymphomas of this type, weakening an argument for a change in classification terminology from DLBCL. Nevertheless, the common morphologic and behavioral features of mouse Burkitt lymphoma and mouse sIg+ “lymphoblastic” tumors suggest that they may share analogous pathogenesis. Therefore, we recommend that the term Burkittlike be applied to mature B-cell neoplasms with proliferative fractions approaching 100% even in the absence of MYC translocation. The term Burkitt lymphoma will be reserved for tumors with compatible morphology, phenotype, and MYC dysregulation because of translocation or genetic manipulation. This area is clearly one in which it can be anticipated that molecular profiling will be of great use in sorting out histologically similar mature B-cell lymphomas.

Finally, although the term plasmablastic lymphoma is used for a variant of human DLBCL, it is subsumed under plasma cell neoplasms in mice (Figure 4). The major reason for this decision is that most mouse plasmacytic lymphomas cover a range of cytologic features, ranging from mature monoclonal plasma cells to plasmablasts in varying proportions. The plasmablastic variant represents one extreme. Human plasmablastic lymphoma, by comparison, typically lacks mature plasma cells and can have a morphologic spectrum that ranges from immunoblastic to Burkitt lymphoma.

Several diseases are recognized as occurring in unusual circumstances. One is found in association with 2 different mutations in a common signaling pathway, and a second is found in response to infection with a unique mixture of MuLVs. These are the diseases of mice homozygous for mutations of Fas or Fasl and mice with the retrovirus-induced immunodeficiency syndrome, MAIDS. These diseases are characterized in their early stages by greatly expanded polyclonal proliferations of lymphocytes that are ultimately replaced by monoclonal populations of B cells. Cells in the early stages of these chronic proliferative disorders do not transplant to immunocompetent or immunodeficient hosts, whereas those cells from late-stage animals can be transferred most readily to mice with impaired immunity. The cells that proliferate in the adoptive hosts are clearly lymphomas, but their presence in the original animal is not often apparent, as the expanded populations of nontransformed cells obscure their presence. The committee was hesitant to classify these lymphomas with those readily diagnosed in the primary host because there was no way to determine the stage of the tumor in the primary host or to know how passage in vivo may have influenced histologic appearance and phenotype. The difficulties associated with defining the MAIDS tumors as true lymphoma have been discussed.26 These disorders and their associations with lymphoma are important areas for further study.

Mice with mutations of Fas or Fasl develop massive lymphadenopathy and splenomegaly due to the polyclonal accumulation of TCRα/β+CD3+CD4CD8B220dull“double-negative” T cells.27 Later, up to 60% of mutants from 6 to 15 months of age have clonal populations of B cells (in spleen or lymph nodes) that are not discernible among the mass of double-negative T cells but can be uncovered by transplantation to immunodeficient hosts.28 The tumors are immunoglobulin class switched, but there are no MYC translocations. They also secrete immunoglobulin in the primary animal, on transplantation, and after adaption to tissue culture. The histologic features on transplantation were described as plasmacytoid lymphoma.28 Some humans with mutations in FAS have developed B-cell lymphomas of diverse types, suggesting analogous processes in mice and humans.29 30 

Mice infected with the LP-BM5 mixture of MuLV develop a degree of splenomegaly and lymphadenopathy rivaled only by mice with mutations in Fas and Fasl.31,32Initially, germinal centers are very prominent with large numbers of plasma cells intermixed with centroblasts and increasing numbers of immunoblasts. The disease is initially polyclonal for both B and T cells,33,34 followed by the appearance of oligoclonal populations of B cells more often than of T cells around 8 weeks after infection.33 By 12 weeks, all mice have monoclonal populations of T or B cells that can be transplanted to immunodeficient hosts.35,36 In occasional mice, lymphomas can result in paralysis and death from invasion of the central nervous system.33 Under the proposed nomenclature, the transplants can be described as DLBCL. B-cell lineage lymphomas occur at high frequencies in patients with genetically determined, iatrogenic, or acquired immunodeficiencies such as AIDS,37 providing possible human parallels.

The content of this publication does not necessarily reflect the views or policies of the Department of Health and Human Services, nor does the mention of trade names, commercial products, or organizations imply endorsement by the United States government.

Supported in part with funds from the Mouse Models of Human Cancers Consortium, National Cancer Institute, National Institutes of Health, and by contract N01-CO56000 from the National Cancer Institute.

H.C.M.III and J.M.W. assisted in the organization of the pathology meeting and prepared the manuscript with assistance from the other authors listed alphabetically. The publication represents the consensus of the committee that included the authors, Robert D. Cardiff, Cory Brayton, James Downing, Hiroshi Hiai, Pier Paolo Pandolfi, Jules J. Berman, Mark S. Tuttle, and Archibald S. Perkins.

The online version of the article contains a data supplement.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

1
Dunn
TB
Normal and pathologic anatomy of the reticular tissue in laboratory mice.
J Natl Cancer Inst.
14
1954
1281
1433
2
Rappaport
H
Tumors of the hematopoietic system
Atlas of Tumor Pathology.
1966
Armed Forces Institute of Pathology
Washington, DC
3
Pattengale
PK
Taylor
CR
Experimental models of lymphoproliferative disease: the mouse as a model for human non-Hodgkin's lymphomas and related leukemias.
Am J Pathol.
113
1983
237
265
4
Lukes
R
Collins
R
Immunological characterization of human malignant lymphomas.
Cancer.
34
1974
1488
1503
5
Fredrickson
TN
Hartley
JW
Morse
HC
III
Chattopadhyay
SK
Lennert
K
Classification of mouse lymphomas.
Curr Top Microbiol Immunol.
194
1994
109
116
6
Lennert
K
Histopathology of Non-Hodgkin's Lymphomas: Based on the Kiel Classification.
1981
Springer-Verlag
New York, NY
7
Stansfeld
AG
Diebold
J
Noel
H
et al
Updated Kiel classification for lymphomas.
Lancet.
1
1988
292
293
8
Fredrickson
TN
Lennert
K
Chattopadhyay
SK
Morse
HC
III
Hartley
JW
Splenic marginal zone lymphomas of mice.
Am J Pathol.
154
1999
805
812
9
Harris
NL
Jaffe
ES
Stein
H
et al
A revised European-American classification of lymphoid neoplasms: a proposal from the International Lymphoma Study Group.
Blood.
84
1994
1361
1392
10
Jaffe
ES
Harris
NL
Stein
H
Vardiman
J
Pathology and Genetics of Tumours of Haematopoietic and Lymphoid Tissues.
2001
IARC Press
Lyons, France
11
Jaffe
ES
Harris
NL
Diebold
J
Muller-Hermelink
H-K
World Health Organization classification of neoplastic diseases of the hematopoietic and lymphoid tissues: a progress report.
Am J Clin Pathol.
111(suppl 1)
1999
S8
S12
12
Morse
HC
III
Qi
C-F
Taddesse-Heath
L
et al
Novel aspects of murine B cell lymphomas.
Curr Top Microbiol Immunol.
246
1999
249
254
13
Taddesse-Heath
L
Morse
HC
III
Lymphoma in genetically engineered mice.
Pathology of Genetically Engineered Mice.
Ward
JM
Mahler
JF
Maronpot
RR
Sundberg
JP
Frederickson
RM
2000
365
381
Iowa State University Press
Ames, IA
14
Hartley
JW
Chattopadhyay
SK
Lander
MR
et al
Accelerated appearance of multiple B cell lymphoma types in NFS/N mice congenic for ecotropic murine leukemia viruses.
Lab Invest.
80
2000
159
169
15
Frith
CG
Wiley
LD
Morphologic classification and correlation of incidence of hyperplastic and neoplastic lesions in mice with age.
J Gerontol.
36
1981
534
545
16
Frith
CH
Ward
JM
Chandra
M
The morphology, immunohistochemistry, and incidence of hematopoietic neoplasms in mice and rats.
Toxicol Pathol.
21
1993
206
218
17
Rowe
WP
Leukemia virus genomes in the chromosomal DNA of the mouse.
Harvey Lect.
71
1978
173
192
18
Rosenberg
N
Jolicoeur
P
Retroviral pathogenesis.
Retroviruses.
Coffin
JM
Hughes
SH
Varmus
HE
1997
475
486
Cold Spring Harbor Laboratory Press
Plainview, NY
19
Ward
JM
Mahler
JF
Maronpot
RR
Sundberg
JP
Frederickson
RM
Pathology of Genetically Engineered Mice.
2000
Iowa State University Press
Ames, IA
20
Steiniger
B
Barth
P
Hellinger
A
The perifollicular and marginal zones of the human splenic white pulp—do fibroblasts guide lymphocyte immigration?
Am J Pathol.
159
2001
501
512
21
Haines
DC
Chattopadhyay
S
Ward
JM
Pathology of aging B6;129 mice.
Toxicol Pathol.
29
2001
653
661
22
Morse
HC
III
Qi
CF
Chattopadhyay
SK
et al
Combined histologic and molecular features reveal previously unappreciated subsets of lymphoma in AKXD recombinant inbred mice.
Leuk Res.
25
2001
719
733
23
Qi
CF
Hori
M
Coleman
AE
et al
Genomic organisation and expression of Bcl6 in murine B-cell lymphomas.
Leuk Res.
24
2000
719
732
24
Ye
BH
Lista
F
Lo Coco
F
et al
Alterations of a zinc finger-encoding gene, BCL-6, in diffuse large-cell lymphoma.
Science.
262
1993
747
750
25
Kovalchuk
AL
Qi
CF
Torrey
TA
et al
Burkitt lymphoma in the mouse.
J Exp Med.
192
2000
1183
1190
26
Morse
HC
III
Hartley
JW
Tang
Y
Chattopadhyay
SK
Giese
N
Fredrickson
TN
Lymphoproliferation as a precursor to neoplasia: what is a lymphoma?
Viruses and Cancer. Cambridge
Minson
A
Neil
J
McCrae
M
1994
265
291
Cambridge University Press
United Kingdom
27
Davidson
WF
Dumont
FJ
Bedigian
HG
Fowlkes
BJ
Morse
HC
III
Phenotypic, functional, and molecular genetic comparisons of the abnormal lymphoid cells of C3H-lpr/lpr and C3H-gld/gld mice.
J Immunol.
136
1986
4075
4084
28
Davidson
WF
Giese
T
Fredrickson
TN
Spontaneous development of plasmacytoid tumors in mice with defective Fas-Fas ligand interactions.
J Exp Med.
187
1998
1825
1838
29
Lim
MS
Straus
SE
Dale
JK
et al
Pathological findings in human autoimmune lymphoproliferative syndrome.
Am J Pathol.
153
1998
1541
1550
30
Straus
SE
Jaffe
ES
Puck
JM
et al
The development of lymphomas in families with autoimmune lymphoproliferative syndrome with germline Fas mutations and defective lymphocyte apoptosis.
Blood.
98
2001
194
200
31
Pattengale
PK
Taylor
CR
Twomey
P
et al
Immunopathology of B-cell lymphomas induced in C57BL/6 mice by dualtropic murine leukemia virus (MuLV).
Am J Pathol.
107
1982
362
377
32
Mosier
DE
Yetter
RA
Morse
HC
III
Retroviral induction of acute lymphoproliferative disease and profound immunosuppression in adult C57BL/6 mice.
J Exp Med.
161
1985
766
784
33
Klinken
SP
Fredrickson
TN
Hartley
JW
Yetter
RA
Morse
HC
III
Evolution of B-cell lineage lymphomas in mice with a retrovirus-induced immunodeficiency syndrome, MAIDS.
J Immunol.
140
1988
1123
1131
34
Klinman
DM
Morse
HC
III
Characteristics of B-cell proliferation and activation in murine AIDS.
J Immunol.
142
1989
1144
1149
35
Tang
Y
Chattopadhyay
SK
Hartley
JW
Fredrickson
TN
Morse
HC
III
Clonal outgrowths of T and B cells in SCID mice reconstituted with cells from mice with MAIDS.
In Vivo.
8
1994
953
960
36
Kubo
Y
Nakagawa
Y
Kakimi
K
et al
Presence of transplantable T-lymphoid cells in C57BL/6 mice infected with murine AIDS virus.
J Virol.
66
1992
5691
5695
37
Ziegler
JL
Beckstead
JA
Volberding
PA
et al
Non-Hodgkin's lymphoma in 90 homosexual men—relation to generalized lymphadenopathy and the acquired immunodeficiency syndrome.
N Engl J Med.
311
1984
565
570
38
Morse
HC
III
Kearney
JF
Isaacson
PG
Carroll
M
Fredrickson
TN
Jaffe
ES
Cells of the marginal zone—origins, function and neoplasia.
Leuk Res.
25
2001
169
178
39
Ward
JM
Taddesse-Heath
L
Perkins
SN
Chattopadhyay
SK
Hursting
SD
Morse
HC
III
Splenic marginal zone B-cell and thymic T-cell lymphomas in p53-deficient mice.
Lab Invest.
79
1999
3
14
40
Enno
A
O'Rourke
JL
Howlett
CR
Jack
A
Dixon
MF
Lee
A
MALToma-like lesions in the murine gastric mucosa after long-term infection with Helicobacter felis: a mouse model of Helicobacter pylori-induced gastric lymphoma.
Am J Pathol.
147
1995
217
222
41
Stansfeld
AG
Low-grade B cell lymphomas.
Lymph Node Biopsy Interpretation. Edinburgh
Stansfeld
AG
1992
229
283
Churchill Livingstone
United Kingdom
42
Fredrickson
TN
Harris
AW
Atlas of Mouse Hematopathology.
2000
Harwood Academic Publishers
Amsterdam, The Netherlands
43
Potter
M
Wiener
F
Plasmacytomagenesis in mice: model of neoplastic development dependent on chromosomal translocations.
Carcinogenesis.
13
1992
1681
1697
44
Kovalchuk
AL
Kim
JS
Park
SS
et al
IL-6 transgenic mouse model for extraosseous plasmacytoma.
Proc Natl Acad Sci U S A.
99
2002
1509
1514
45
Lu
L-M
Hiai
H
Mixed phenotype lymphomas in thymectomized (SL/Kh × AKR/Ms)F1 mice.
Jpn J Cancer Res.
90
1999
1218
1223
46
Fehniger
TA
Suzuki
K
Ponnappan
A
et al
Fatal leukemia in interleukin 15 transgenic mice follows early expansions in natural killer and memory phenotype CD8+ T cells.
J Exp Med.
193
2001
219
231
47
Baldassarre
G
Fedele
M
Battista
S
et al
Onset of natural killer cell lymphomas in transgenic mice carrying a truncated Hmgi-C gene by the chronic stimulation of the IL-2 and IL-5 pathway.
Proc Natl Acad Sci U S A.
98
2001
7970
7975
48
Kogan
SC
Ward
JM
Anver
MR
et al
Bethesda Proposals for Classification of Nonlymphoid Hematopoietic Neoplasms in Mice.
Blood.
100
2002
238
245

Author notes

Herbert C. Morse III, 7 Center Dr, Rm 7/304, MSC 0760, Bethesda, MD 20892-0760; e-mail: hmorse@niaid.nih.gov.

Sign in via your Institution